Next Article in Journal
Design and Synthesis of Pyridine-Based Pyrrolo[2,3-d]pyrimidine Analogs as CSF1R Inhibitors: Molecular Hybridization and Scaffold Hopping Approach
Previous Article in Journal
Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets
Previous Article in Special Issue
Hormonal Treatment of Endometriosis: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches

1
Research Institute for Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
2
Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
3
Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea
4
Department of Obstetrics and Gynecology, School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea
5
Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this study.
Pharmaceuticals 2025, 18(6), 813; https://doi.org/10.3390/ph18060813
Submission received: 15 April 2025 / Revised: 19 May 2025 / Accepted: 27 May 2025 / Published: 28 May 2025
(This article belongs to the Special Issue Pharmacotherapy of Endometriosis)

Abstract

:
Endometriosis is a chronic gynecological disorder characterized by the presence of endometrial-like tissue outside the uterus, leading to inflammation, pain, and infertility. Emerging evidence indicates that endometriotic lesions exhibit cancer-like properties, including metabolic reprogramming marked by increased glucose uptake, enhanced Warburg’s effect, and altered mitochondrial function. These metabolic adaptations support cell survival under hypoxic conditions and contribute to immune evasion and sustained proliferation. This review summarizes current findings on the molecular mechanisms driving metabolic reprogramming in endometriosis, including the roles of mitochondrial dysfunction, hypoxia-inducible factor (HIF) signaling, the PI3K/AKT/mTOR pathway, inflammatory cytokines, and genetic and epigenetic regulators. In addition, we discuss therapeutic strategies targeting glycolytic pathways using both synthetic inhibitors and natural compounds, which represent promising non-hormonal options. Finally, we highlight the need for further preclinical and clinical studies to validate metabolic interventions and improve outcomes for patients with endometriosis.

1. Introduction

Endometriosis is a chronic gynecological condition characterized by the presence of endometrial-like tissue outside the uterus, mainly in the ovaries, fallopian tubes, and peritoneum [1]. This ectopic endometrium undergoes cyclic changes similar to the normal endometrium, causing inflammation, fibrosis, and adhesion formation. Endometriosis is estimated to affect 10–15% of reproductive age women with severe health problems, such as chronic pelvic pain, dysmenorrhea, dyspareunia, and infertility [2,3]. Despite its high prevalence, the precise pathogenesis of this disease has not yet been fully elucidated [4,5]. Several factors, including hormonal imbalance, immune dysfunction, genetic predisposition, and environmental influences, contribute to the development and progression of this disease. Due to the non-specific symptoms and limited public recognition, many patients experience delays in diagnosis and treatment. Current treatment options involve surgical and hormonal therapies, which focus on symptom management rather than fundamental treatment [6,7]. Since hormone therapy is often accompanied by serious side effects, the need for non-hormonal treatments has been in the spotlight [8].
Endometriosis manifests several cancer-like characteristics, including local invasion, distant metastasis, apoptotic cell death resistance, and immune evasion [9]. Recent evidence shed light on shared pathological mechanisms of the two diseases; a process known as metabolic reprogramming is one of the key mechanisms. Specifically, endometriotic lesions also exhibit a metabolic profile similar to the Warburg’s effect in cancer cells, which is characterized by increased glucose uptake, enhanced glycolysis, and altered mitochondrial function [10]. The metabolic shift to aerobic glycolysis promotes energy production and the biosynthesis of cellular building blocks, which helps the survival and proliferation of ectopic endometrial cells [11]. The Warburg’s effect enables migrated endometrial cells to adapt to the low-oxygen environment of the peritoneal cavity, avoiding cell death and maintaining an inflammatory condition. Therefore, targeting glycolytic metabolism—a key pathway underlying the Warburg’s effect—emerges as a promising therapeutic strategy for endometriosis.
This review provides a detailed overview of the molecular mechanisms underlying aerobic glycolysis in endometriosis, along with therapeutic approaches, including metabolic inhibitors and dietary interventions. A comprehensive understanding of these metabolic features is expected to facilitate the development of more effective treatment strategies and ultimately improve clinical outcomes.

2. Overview of the Warburg’s Effect

The Warburg’s effect, first described by German physician and biochemist Otto Warburg in 1923, describes the metabolic reprogramming of cancer cells in which glucose metabolism is switched from oxidative phosphorylation to aerobic glycolysis, even when sufficient oxygen is present [12]. Normal cells produce ATP mainly through mitochondrial oxidative phosphorylation, while cancer cells prefer glycolysis and convert pyruvate to lactic acid without entering the tricarboxylic acid (TCA) cycle. This phenomenon provides a number of benefits that promote tumor growth, including rapid production of ATP, availability for biosynthetic building blocks, and adaptation to hypoxic environments [13]. It was selected as one of the 10 hallmarks of cancer in Douglas Hanahan’s landmark article “Hallmarks of Cancer: The Next Generation” in 2011 [14].
As illustrated in Figure 1, the Warburg’s effect is facilitated at the molecular level by the upregulation of key glycolytic enzymes and the activation of oncogenic signaling pathways. Many cancer cells overexpress hexokinase 2 (HK2), which catalyzes the first step of glycolysis by phosphorylating glucose and trapping it in the cell [15]. Another key regulator is pyruvate kinase M2 (PKM2), which is an isozyme of the less active pyruvate kinase that exists in a dimeric form in cancer cells [16]. This altered enzyme activity slows down the conversion of phosphoenolpyruvic acid to pyruvic acid, resulting in the accumulation of intermediates that serve as precursors for anabolic pathways involved in nucleotide, amino acid, and lipid biosynthesis. In addition, increased expression of lactate dehydrogenase A (LDHA) promotes the conversion of pyruvate to lactate, facilitating a continuous flow of glycolysis by regenerating NAD⁺. The Warburg’s effect is closely related to tumor-promoting signaling pathways that regulate metabolism [17]. An essential transcription factor, hypoxia-inducible factor-1α (HIF-1α) is stabilized in hypoxic environments but is upregulated in many cancers regardless of oxygen availability. HIF-1α induces the expression of various glycolysis-related proteins, including glucose transporters (GLUT1 and GLUT3), and pyruvate dehydrogenase kinase 1 (PDK1) and 3 (PDK3). PDK inhibits the activity of pyruvate dehydrogenase (PDH), thereby preventing pyruvate from being converted to acetyl-CoA and entering the TCA cycle. Another important signaling cascade is the PI3K/AKT/mTOR pathway [18]. This pathway upregulates GLUT1 and HK2 to enhance glucose uptake and glycolysis while inducing cell survival and proliferation. Furthermore, the oncogene MYC plays an important role in enhancing the metabolic reprogramming of cancer cells by activating the production of glycolytic enzymes and mitochondrial biogenesis [19].
The Warburg’s effect is caused by both active oncogenes and the suppression of tumor suppressor genes. The tumor suppressor gene p53 is frequently mutated in cancer and plays an important role in metabolic regulation [20,21]. Wild-type p53 negatively regulates glycolysis by inducing TP53-induced glycolysis and apoptosis regulator (TIGAR), which lowers the level of fructose-2,6-phosphate, a key activator of phosphofructokinase-1 (PFK1) [22]. p53 also enhances mitochondrial respiration by promoting the expression of cytochrome c oxidase 2 (SCO2), an important component of the electron transport chain [23]. When the p53 function is lost in cancer cells, these constraints disappear, allowing glycolysis to proceed unchecked and metabolic adaptation to be further enhanced.
The Warburg’s effect is often related to mitochondrial dysfunction, although cancer cells do not completely abandon oxidative phosphorylation [24]. Instead, many tumors exhibit metabolic plasticity, utilizing both glycolysis and mitochondrial metabolism, depending on the environmental conditions [25]. This flexibility allows cancer cells to survive in fluctuating oxygen levels and nutrient availability. Also, glutamine metabolism is often increased in tumors, supplying α-ketoglutarate to the TCA cycle and supporting the biosynthesis pathways necessary for rapid proliferation [26]. The interplay between these processes and alternative metabolic pathways highlights the complexity of cancer metabolism that extends well beyond energy production to include cell signaling, redox balance, and immune evasion.
Metabolic reprogramming observed in cancer cells has a significant impact on tumor progression. Increased glucose uptake and lactate production promote invasion and metastasis while contributing to the formation of an acidic microenvironment that suppresses immune cell activity [27,28]. In addition, the conversion to aerobic glycolysis enables the rapid production of macromolecules required for cell division, which is beneficial for cancer cell growth [29]. Moreover, it can produce ATP quickly without the increase in reactive oxygen species (ROS) that inevitably occurs in mitochondrial respiration using oxygen. The ability of cancer cells to re-adjust their metabolism as described above enables them to resist apoptosis and evade immune surveillance, which increases the complexity of treatment strategies by causing resistance not only to conventional chemotherapeutics but also immunotherapies.
Understanding the molecular mechanisms underlying the Warburg’s effect can provide valuable insights into the metabolic vulnerability of cancer cells. Aerobic glycolysis is a hallmark of tumor metabolism; however, the extensive metabolic network that supports tumor growth is highly dynamic, influenced by genetic mutations, tumor microenvironmental factors, and immune interactions. Unraveling these complex metabolic dependencies presents potential opportunities for cancer therapy.

3. Metabolic Reprogramming in Endometriosis

Recent research has highlighted metabolic reprogramming as a key factor in the pathophysiology of endometriosis, influencing cell survival, proliferation, and immune evasion [10,30,31,32]. In normal endometrial cells, the metabolic process is tightly regulated by the hormonal cycle. However, ectopically located endometriotic cells in the peritoneal cavity exhibit distinct metabolic alterations that distinguish them from normal cells (Figure 2) [33,34]. A characteristic of this change is the shift to aerobic glycolysis, commonly known as the Warburg’s effect, in which the pathway is re-routed so that glucose metabolism prefers glycolysis rather than oxidative phosphorylation (OXPHOS). Increases in lactic acid production lead to an acidic microenvironment that promotes inflammation and immune evasion. In addition, fatty acid oxidation is increased in endometriotic lesions, providing an alternative energy source to maintain cell survival under stress conditions [35]. Glutamine metabolism has also been reported to contribute to the production of NADPH, which is essential for cell proliferation, and to the biosynthesis process [30,36]. Hypoxia, a common feature of endometriotic lesions, plays an important role in promoting these metabolic changes [37,38]. Since the intraperitoneal cavity, where endometriosis develops, has a lower oxygen concentration than normal tissues, HIF-1α is activated under hypoxic conditions, which, in turn, inhibits mitochondrial oxidative phosphorylation and enhances gene expression of the glycolysis pathway. As a result, mitochondrial dysfunction—characterized by decreased efficiency of the electron transport chain, increased ROS production, and mitochondrial DNA mutations—is frequently observed in endometriosis cells. These changes contribute to the cellular stress response, further enhancing inflammation and disease progression [39]. Initially, this change was understood to be simply an adaptation to the intraperitoneal environment with a shortage of oxygen; however, a series of studies conducted in the late 2010s showed that cancer-like genetic mutations appear in endometrial tissue and accumulate further in ectopic endometrial tissue [40,41]. Therefore, it has been proposed that the accumulation of these genetic mutations may lead to metabolic abnormalities in endometriotic cells.
Metabolic reprogramming in endometriosis, as represented in Figure 2, represents a multifaceted process involving increased glycolysis, enhanced fatty acid oxidation, and mitochondrial dysfunction, all contributing to the persistence and progression of the disease [42]. Understanding these metabolic adaptations may open new avenues for potential therapeutic interventions targeting specific metabolic pathways to alleviate the burden of endometriosis.

4. Molecular Mechanisms Underlying Warburg-like Metabolic Reprogramming

4.1. Hypoxia-Inducible Factor (HIF) Signaling and Glycolysis Activation

Hypoxia is a characteristic feature of endometriotic lesions, which is caused by the nature of the peritoneal environment, insufficient vascularization of ectopic endometrial tissue, and high metabolic demand [37]. Under these conditions, HIF-1α is stabilized and translocated to the nucleus, likely avoiding proteasome degradation. HIF-1α activates the transcription of genes essential for survival and glycolysis under oxygen deprivation. McKinnon et al. explained that this activation increases the expression of glucose transporters such as GLUT1 and GLUT4, enabling a continuous supply of glucose (Figure 3) [43]. HIF-1α also increases the expression of important glycolytic enzymes, including HK2, PFK, PDK1, and LDHA [44]. Several researchers have further emphasized that TGF-β1 and hypoxia increase the activity of HIF-1α, leading to the enhancement of the process [10,45,46,47]. In addition, hypoxia-induced stabilization of HIF-2α and its interaction with the VEGF pathway promote angiogenesis and establish a positive feedback loop that exacerbates hypoxic conditions and enhances pathological processes such as neovascularization and tissue invasion [48,49]. Recently, non-canonical mechanisms have been proposed in which HIF is activated by metabolic products such as lactate and succinate even when the tissue is not actually hypoxic, indicating that these mechanisms may also affect endometriosis [50]. Prior research showed that the buildup of ROS completes the pathological cycle and supports the growth of the lesion by stabilizing HIF-1α and promoting inflammation [51,52,53]. In particular, oxidative stress and reactive oxygen species (ROS) production are both a consequence and a driving force of glycolytic reprogramming. Accumulated ROS can activate key signaling pathways such as HIF-1α and NF-κB, which, in turn, enhance the expression of glycolytic enzymes and glucose transporters, thereby reinforcing the glycolytic phenotype [54,55]. Conversely, the shift toward aerobic glycolysis reduces mitochondrial oxidative phosphorylation, which can lead to electron leakage and further ROS generation [56]. This bidirectional relationship contributes to a self-sustaining cycle that supports the survival and proliferation of endometriotic cells under hypoxic conditions.

4.2. PI3K/AKT/mTOR Pathway and Metabolic Changes

A number of studies have shown that the PI3K/AKT/mTOR pathway is deeply involved in the metabolic reprogramming of endometriosis [57,58]. As presented in Figure 3, activation of PI3K induces downstream phosphorylation of AKT and activation of mTORC1, which, in turn, enhances HIF1A expression and glycolytic enzyme activity. The mTORC1 complex not only promotes glycolysis but also protein synthesis, lipid biosynthesis, and nucleotide production to meet the metabolic demands of rapidly dividing cells. Crosstalk with AMP-activated protein kinase (AMPK) and feedback regulation through Tuberous sclerosis complex 2 (TSC2) ensure metabolic balance, whereas persistent mTOR activation in endometriotic lesions results in uncontrolled glycolysis and cell proliferation [59]. Additional evidence from protein and transcriptome studies demonstrates that phosphatase and tensin homolog deleted on chromosome 10 (PTEN) loss and AKT hyperactivation lead to metabolic rearrangements that contribute to extracellular matrix remodeling, angiogenesis, and invasiveness [60,61,62]. Furthermore, PIK3CA may be involved in encouraging the glycolysis of endometriosis, because it is one of the cancer-like mutations discovered in endometriotic tissue [63].

4.3. Role of Inflammatory Cytokines and Metabolic Changes

Chronic inflammation is a distinctive feature of endometriosis, and inflammatory cytokines directly affect cellular metabolism. Young et al. (2014, 2016) demonstrated that transforming growth factor-β1 (TGF-β1) regulates inhibitor of DNA-binding 2 (ID2), which, in turn, releases HIF-1α from inhibition and enhances the process [10,64]. In addition, cytokines such as interleukin (IL)-6, IL-1β, and tumor necrosis factor-α (TNF-α) activate the nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways, which upregulate glycolytic enzymes and downregulate mitochondrial oxidative phosphorylation (Figure 3) [65,66]. Several studies have confirmed that inflammatory cytokines promote glucose uptake and lactate production in endometriotic cells [67,68]. The studies also revealed that chemokines such as CXCL12 and its receptor CXCR4 play an important role in regulating glucose metabolism and cell migration [69,70]. Furthermore, several investigations have provided transcriptomic and proteomic evidence that inflammatory signals induce metabolic adaptation by inhibiting mitochondrial oxidative phosphorylation while simultaneously upregulating certain mitochondrial and glycolytic enzymes involved in alternative metabolic routes [71,72]. This seemingly paradoxical regulation reflects a cellular compensatory mechanism; while mitochondrial respiration is suppressed due to hypoxia or inflammation-induced damage, cells increase the expression of enzymes related to mitochondrial biogenesis, antioxidant defense (e.g., SOD2), or metabolic flexibility (e.g., citrate synthase and PDK1) to adapt to stress and sustain energy production through glycolysis and other non-canonical pathways. Thus, the upregulation of these enzymes does not necessarily indicate enhanced mitochondrial activity but rather a shift in metabolic programming to cope with mitochondrial dysfunction [73,74,75].

4.4. Genetic and Epigenetic Factors Contributing to Metabolic Reprogramming

Epigenetic regulation and genetic predisposition contribute significantly to metabolic rearrangement in endometriosis. According to whole-genome studies and epigenomic profiling, endometriosis lesions adapt through stable genetic and epigenetic modifications [76,77,78]. Genomic modifications such as single-nucleotide polymorphisms in metabolic genes and mutations in regulators of the PI3K/AKT/mTOR pathway can induce metabolic reprogramming. Several studies have emphasized that DNA methylation changes, histone modifications, and regulation by microRNAs such as miR-21, miR-210, and miR-145 affect the pathways and mitochondrial function (Figure 3) [79,80,81]. In addition to miRNAs, it has been found that some lncRNAs, such as H19 and MALAT1, and some circRNAs play an important role in the regulation of glycolysis [82,83,84,85]. Wen et al. [86] highlighted key lncRNAs that promote the expression of glycolytic enzymes and affect glucose metabolism in endometriotic cells.

4.5. The Role of Mitochondrial Dysfunction in the Progression of Endometriosis

Mitochondrial dysfunction is an important factor in the metabolic reprogramming of endometriotic cells. Several researchers have demonstrated that the reduction of prohibitin (PHB) and mitochondrial dysfunction increases the dependence on metabolic reprogramming (Figure 3) [87,88,89]. In addition, proteomic changes were identified that are consistent with mitochondrial dysfunction. For instance, mitochondrial dysfunction is associated with the downregulated expression of key enzymes in the oxidative phosphorylation pathway [90,91,92]. The increase in ROS generated by dysfunctional mitochondria contributes to genomic instability and chronic inflammation. Moreover, mitochondrial dysfunction has been linked to endometriotic cell proliferation and invasion of the surrounding tissues through ROS-stabilized HIF-1α [93,94].

4.6. Interaction with Angiogenesis and Matrix Remodeling

Angiogenesis and remodeling of the extracellular matrix (ECM) are closely related to metabolic changes (Figure 3). Increased lactate production and secretion promote VEGF expression and neovascularization [95,96]. Several studies have reported that increased matrix metalloproteinases (MMPs), by the glycolysis signal, allows the invasion into neighboring tissues [97,98].

5. Current Research on Glycolysis Inhibitors in Endometriosis

As shown in Table 1 and illustrated in Figure 4, ongoing research on glycolysis inhibitors for endometriosis is being conducted using various compounds, depending on the molecular target. A brief comparison of key candidate compounds is summarized in Table 2. The PDK family has emerged as the most important target, and dicarboxylate (DCA), a drug that was long ago approved by the FDA as an inhibitor of lactic acidosis, is the most extensively studied candidate. In previous preclinical and early clinical studies (Horne et al. 2019; Lee HC et al. 2019; Leow HW et al. 2021), DCA was shown to restore mitochondrial function by inhibiting PDK1, reduce lactic acid production, and alleviate endometriosis-related pain [99,100,101]. Similarly, Caesalpinia sappan extract was shown to inhibit the glycolytic activity of endometriotic cells and induce apoptosis by reducing the expression of PDK1 and PDK3 [102]. However, it should be noted that DCA requires relatively high concentrations (in the millimolar range) to exert its effects, which raises concerns about specificity and potential off-target effects. Despite this limitation, its progression to early-phase clinical trials highlights its significance in the field and underscores the need for further research into more potent and selective metabolic inhibitors. Moreover, enzymes like LDHA and PKM2 are garnering attention [103]. LDHA inhibition through shRNA has been shown to suppress glycolysis and promote cell death in endometriotic cells [104]. In addition, Cho et al. (2022) reported that Prunella vulgaris extract effectively reduces aerobic glycolysis and prevents lesion growth by inhibiting LDHA as the main target and inhibiting the expression of PDK1 and PDK3 at the same time [105]. They also showed that ursolic acid, a constituent of Prunella vulgaris, effectively inhibits LDHA [106]. Recently, a number of research groups have been actively conducting research on LDHA inhibitors with anticancer activity, so it is expected that research on these drugs for endometriosis will also be expanded. Research on PKM2 has revealed that inhibiting PKM2 using succinic acid reduces cell viability by inhibiting the corresponding process in endometriosis stromal cells [107]. Proviral insertion in murine lymphomas 2 (PIM2) kinase has been identified as a key regulator of the expression of glycolysis enzymes such as PKM2 and HK2, while inhibiting PIM2 by treating SMI-4a reduces glycolysis and fibrosis [108,109]. In addition, p21-activated kinase 5 (PAK5) increases the protein stability of PKM2; thus, inhibiting PAK5 with GNE 2661 inhibited glycolysis and growth in endometriosis [110].
Another notable target is phosphofructokinase/fructose bisphosphatase (PFKFB3), and the inhibitor PFK-015 has been shown to inhibit cell proliferation, migration, and invasion by suppressing the glycolysis process through the inhibition of the enzyme at the cellular and in vivo levels [117]. Recent studies have confirmed that heat shock factor 1 (HSF1) regulates the expression of PFKFB3 and that the inhibitor KRIBB11 suppresses endometriosis in animal experiments [113,114]. Glucose analog 2-DG inhibits HK2, the most upstream enzyme of the glycolysis pathway. It was verified that 2-DG suppressed the infiltration of differentiated M2 macrophages in the endometriotic lesions [121]. In the study, it was found that the Mettl3/Trib1/ERK-STAT3 pathway plays an important role [116].
The involvement of Aurora kinase A (AURKA) and carboxyl terminus of the Hsc70-interacting protein (CHIP) pathways in glycolysis regulation provides new therapeutic targets. Alisertib (AURKA inhibitor) and YL-109 (CHIP agonist) have shown potential for the treatment of endometriosis in animal studies [115,118]. Resveratrol, a natural product found in grapes, has demonstrated inhibitory effects on glycolysis and anti-angiogenesis in animal models when administered alone or in combination with atorvastatin [112]. Gui-Zhi-Fu-Ling capsules, a traditional Chinese medicine, have also shown a positive effect on endometriosis by inhibiting multiple targets such as TGF-β1, GLUT4, and VEGF [111,122].
However, despite the accumulation of research on the metabolic abnormality of endometriosis, especially aerobic glycolysis, more preclinical and clinical research needs to be carried out. Most animal experiments using molecular targeted inhibitors are still not enough to be considered as accurate preclinical research. To date, the only clinical trial targeting the glycolytic pathway of endometriosis has found that DCA could suppress pain in endometriosis patients. We expect that research on drug development using diverse molecular targets in this field will become increasingly active in the future.

6. Clinical Implications of Metabolic Reprogramming in Endometriosis

Metabolic reprogramming, especially the shift to glycolysis, plays an important role in the persistence and progression of endometriotic lesions. Increased lactate production and altered energy metabolism create an acidic microenvironment that promotes angiogenesis, tissue invasion, and immune evasion [119,120]. These metabolic abnormalities lead to changes in the extracellular matrix composition and the immune cell infiltration. This metabolic state not only helps the survival of ectopic endometrial cells but also promotes chronic inflammation, which increases the severity of symptoms (Figure 5). The continuous metabolic flux can alter the local nerve fiber density and contribute to the development of neuropathic pain [10,123]. In particular, pain is the most critical symptom that reduces the quality of life of patients with endometriosis. In clinical studies, PDK1 inhibitor DCA has been confirmed to contribute to the reduction of pain in endometriosis patients.
Inflammatory responses affecting the oocyte, the endometrium, and the hormonal regulation are closely associated with infertility in endometriosis patients [124]. High lactate levels and oxidative stress can reduce the quality of eggs, interfere with the receptivity of the endometrium, and affect embryo implantation [125]. Metabolic environment change can disrupt hormonal signaling in reproductive tissues and cause oxidative DNA damage [126,127]. In addition, the accumulation of inflammatory cytokines and neurovascular factors in the active lesion of glycolysis can cause chronic pelvic pain and dysmenorrhea [128]. The pain pathway is further sensitized by local nerve growth, cytokine release, and metabolic byproducts such as lactic acid and reactive oxygen species. Fibrosis is promoted by altered metabolites, and inflammatory cytokines induce serious complications, including adhesions to the surrounding tissue of endometriosis lesions.
Currently, the standard drug treatment for endometriosis, hormonal therapy (e.g., GnRH agonists and oral contraceptives), aims to suppress estrogen-induced proliferation, although it does not directly act on metabolic changes (Figure 5) [129,130]. Recent evidence suggests that the combination of hormonal therapy and metabolic interventions may improve treatment outcomes. For example, mTOR inhibitors and glycolysis inhibitors can improve the effectiveness of standard treatments by addressing the metabolic dependency of endometriosis lesions. Metabolic therapy can help overcome treatment resistance in patients with active metabolic lesions. Furthermore, anti-inflammatory and mitochondrial-targeted therapies can complement existing therapies to reduce recurrence rates and provide more effective symptom relief. However, it is worth noting that most of the supporting studies for these metabolic interventions are limited to preclinical research, with only one small-scale clinical trial conducted to date. As such, the current evidence base remains insufficient, underscoring the need for further clinical investigations to validate these promising therapeutic strategies.

7. Conclusions and Future Perspectives

Warburg-like metabolic reprogramming is one of the fundamental features of endometriosis, contributing to lesion survival, progression, and symptom severity. Under hypoxic and inflammatory conditions, the transition to this process promotes angiogenesis, immune evasion, and tissue invasion while also exacerbating pain and infertility. Understanding these metabolic changes has led to new treatment methods that target metabolic vulnerabilities in endometriotic lesions, as well as hormonal pathways. It is expected that metabolic inhibitors, when combined with existing treatments, will enhance treatment efficacy, reduce recurrence, and improve patients’ quality of life. As research continues, we believe that metabolic profiling and precision medicine approaches will play an increasingly important role in the clinical management of endometriosis.
Future molecular studies of the Warburg’s effect in endometriosis should focus on several key areas. First, the precise regulatory mechanisms of HIF-1α and HIF-2α activation under fluctuating hypoxic conditions in the endometriotic microenvironment should be elucidated. It is important to understand how these factors interact with inflammatory cytokines and hormonal signaling. Second, the role of mitochondrial dynamics, including mitophagy and biogenesis, in glycolytic reprogramming maintenance requires further investigation. Third, studies should explore the post-transcriptional regulation of glycolytic enzymes and transporters, with a focus on non-coding RNAs such as lncRNAs and circRNAs. Fourth, the development of advanced metabolomics and single-cell transcriptomics technologies will enable the identification of subpopulations of endometriotic cells with distinct metabolic phenotypes, which will allow for personalized treatment. Finally, large-scale translational studies are needed to validate the efficacy of metabolic biomarkers for early diagnosis, treatment monitoring, and prognosis prediction.
This area of research will help to deepen our understanding of metabolic changes and develop new therapeutic strategies for endometriosis. Future studies will likely explore the use of combination metabolic inhibitors and non-hormonal therapies in patient populations that do not respond to or are contraindicated for hormonal therapy. Clinical trials focused on metabolic regulation are poised to change the landscape of endometriosis treatment and have the potential to improve long-term outcomes and quality of life for affected women.

Author Contributions

Conceptualization, B.-S.K., B.K., W.K. and K.-T.H.; writing—original draft preparation, B.-S.K., B.K. and S.Y.; writing—review and editing, W.P., S.-J.B., J.J., W.K. and K.-T.H.; visualization, B.-S.K. and B.K.; supervision, K.-T.H.; project administration, W.K. and K.-T.H.; funding acquisition, K.-T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the National Research Foundation of Korea (NRF) grants funded by the Korean government (MIST) (grant no. RS-2022-NR070154).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Giudice, L.C. Clinical practice. Endometriosis. N. Engl. J. Med. 2010, 362, 2389–2398. [Google Scholar] [CrossRef] [PubMed]
  2. Simoens, S.; Dunselman, G.; Dirksen, C.; Hummelshoj, L.; Bokor, A.; Brandes, I.; Brodszky, V.; Canis, M.; Colombo, G.L.; DeLeire, T.; et al. The burden of endometriosis: Costs and quality of life of women with endometriosis and treated in referral centres. Hum. Reprod. 2012, 27, 1292–1299. [Google Scholar] [CrossRef]
  3. World Health Organization (WHO). Endometriosis. Available online: https://www.who.int/news-room/fact-sheets/detail/endometriosis (accessed on 26 May 2025).
  4. Mehedintu, C.; Plotogea, M.N.; Ionescu, S.; Antonovici, M. Endometriosis still a challenge. J. Med. Life 2014, 7, 349–357. [Google Scholar] [PubMed]
  5. Ping, S.; Ma, C.; Liu, P.; Yang, L.; Yang, X.; Wu, Q.; Zhao, X.; Gong, B. Molecular mechanisms underlying endometriosis pathogenesis revealed by bioinformatics analysis of microarray data. Arch. Gynecol. Obstet. 2016, 293, 797–804. [Google Scholar] [CrossRef]
  6. Ferrero, S.; Evangelisti, G.; Barra, F. Current and emerging treatment options for endometriosis. Expert Opin. Pharmacother. 2018, 19, 1109–1125. [Google Scholar] [CrossRef]
  7. Guo, S.W. Recurrence of endometriosis and its control. Hum. Reprod. Update 2009, 15, 441–461. [Google Scholar] [CrossRef] [PubMed]
  8. Hughes, C.L.; Foster, W.G.; Agarwal, S.K. The Impact of Endometriosis across the Lifespan of Women: Foreseeable Research and Therapeutic Prospects. BioMed Res. Int. 2015, 2015, 158490. [Google Scholar] [CrossRef] [PubMed]
  9. Thomas, E.J.; Campbell, I.G. Evidence that endometriosis behaves in a malignant manner. Gynecol. Obstet. Investig. 2000, 50 (Suppl. S1), 2–10. [Google Scholar] [CrossRef]
  10. Young, V.J.; Brown, J.K.; Maybin, J.; Saunders, P.T.; Duncan, W.C.; Horne, A.W. Transforming growth factor-beta induced Warburg-like metabolic reprogramming may underpin the development of peritoneal endometriosis. J. Clin. Endocrinol. Metab. 2014, 99, 3450–3459. [Google Scholar] [CrossRef]
  11. Hou, S.; Lei, S.; Peng, H.; Weng, L.; Lv, S.; Li, M.; Zhao, D. Downregulating HK2 inhibits proliferation of endometrial stromal cells through a noncanonical pathway involving phosphorylation of signal transducer and activator of transcription 1 in endometriosis. Biol. Reprod. 2022, 107, 488–499. [Google Scholar] [CrossRef]
  12. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  13. Liberti, M.V.; Locasale, J.W. The Warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef]
  14. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  15. Mathupala, S.P.; Ko, Y.H.; Pedersen, P.L. Hexokinase-2 bound to mitochondria: Cancer’s stygian link to the “Warburg Effect” and a pivotal target for effective therapy. Semin. Cancer Biol. 2009, 19, 17–24. [Google Scholar] [CrossRef]
  16. Zahra, K.; Dey, T.; Ashish; Mishra, S.P.; Pandey, U. Pyruvate Kinase M2 and Cancer: The Role of PKM2 in Promoting Tumorigenesis. Front. Oncol. 2020, 10, 159. [Google Scholar] [CrossRef]
  17. Courtnay, R.; Ngo, D.C.; Malik, N.; Ververis, K.; Tortorella, S.M.; Karagiannis, T.C. Cancer metabolism and the Warburg effect: The role of HIF-1 and PI3K. Mol. Biol. Rep. 2015, 42, 841–851. [Google Scholar] [CrossRef] [PubMed]
  18. Hoxhaj, G.; Manning, B.D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef]
  19. Dong, Y.; Tu, R.; Liu, H.; Qing, G. Regulation of cancer cell metabolism: Oncogenic MYC in the driver’s seat. Signal Transduct. Target. Ther. 2020, 5, 124. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, C.; Liu, J.; Liang, Y.; Wu, R.; Zhao, Y.; Hong, X.; Lin, M.; Yu, H.; Liu, L.; Levine, A.J.; et al. Tumour-associated mutant p53 drives the Warburg effect. Nat. Commun. 2013, 4, 2935. [Google Scholar] [CrossRef]
  21. Liang, Y.; Liu, J.; Feng, Z. The regulation of cellular metabolism by tumor suppressor p53. Cell Biosci. 2013, 3, 9. [Google Scholar] [CrossRef]
  22. Lee, P.; Vousden, K.H.; Cheung, E.C. TIGAR, TIGAR, burning bright. Cancer Metab. 2014, 2, 1. [Google Scholar] [CrossRef]
  23. Zhang, X.D.; Qin, Z.H.; Wang, J. The role of p53 in cell metabolism. Acta Pharmacol. Sin. 2010, 31, 1208–1212. [Google Scholar] [CrossRef] [PubMed]
  24. Zong, W.X.; Rabinowitz, J.D.; White, E. Mitochondria and Cancer. Mol. Cell 2016, 61, 667–676. [Google Scholar] [CrossRef] [PubMed]
  25. Fendt, S.M.; Frezza, C.; Erez, A. Targeting Metabolic Plasticity and Flexibility Dynamics for Cancer Therapy. Cancer Discov. 2020, 10, 1797–1807. [Google Scholar] [CrossRef]
  26. Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef]
  27. Arner, E.N.; Rathmell, J.C. Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell 2023, 41, 421–433. [Google Scholar] [CrossRef] [PubMed]
  28. Ohshima, K.; Morii, E. Metabolic Reprogramming of Cancer Cells during Tumor Progression and Metastasis. Metabolites 2021, 11, 28. [Google Scholar] [CrossRef]
  29. Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
  30. Sarsenova, M.; Lawarde, A.; Pathare, A.D.S.; Saare, M.; Modhukur, V.; Soplepmann, P.; Terasmaa, A.; Kaambre, T.; Gemzell-Danielsson, K.; Lalitkumar, P.G.L.; et al. Endometriotic lesions exhibit distinct metabolic signature compared to paired eutopic endometrium at the single-cell level. Commun. Biol. 2024, 7, 1026. [Google Scholar] [CrossRef]
  31. Chen, Q.; Jiao, Y.; Yin, Z.; Fu, X.; Guo, S.; Zhou, Y.; Wang, Y. Establishment of a novel glycolysis-immune-related diagnosis gene signature for endometriosis by machine learning. J. Assist. Reprod. Genet. 2023, 40, 1147–1161. [Google Scholar] [CrossRef]
  32. Guo, S.; Chen, Q.; Liang, J.; Wu, H.; Li, L.; Wang, Y. Correlation of Glycolysis-immune-related Genes in the Follicular Microenvironment of Endometriosis Patients with ART Outcomes. Reprod. Sci. 2024, 31, 3357–3367. [Google Scholar] [CrossRef] [PubMed]
  33. Toniyan, K.A.; Malkov, A.A.; Biryukov, N.S.; Gorbacheva, E.Y.; Boyarintsev, V.V.; Ogneva, I.V. The Cellular Respiration of Endometrial Biopsies from Patients with Various Forms of Endometriosis. Int. J. Mol. Sci. 2024, 25, 3680. [Google Scholar] [CrossRef] [PubMed]
  34. Khashchenko, E.P.; Vysokikh, M.Y.; Marey, M.V.; Sidorova, K.O.; Manukhova, L.A.; Shkavro, N.N.; Uvarova, E.V.; Chuprynin, V.D.; Fatkhudinov, T.K.; Adamyan, L.V.; et al. Altered Glycolysis, Mitochondrial Biogenesis, Autophagy and Apoptosis in Peritoneal Endometriosis in Adolescents. Int. J. Mol. Sci. 2024, 25, 4238. [Google Scholar] [CrossRef]
  35. Tu, J.L.; Fang, R.X. Identification of fatty acid metabolism hub genes in endometriosis using integrative bioinformatics analysis. Front. Med. 2025, 12, 1529074. [Google Scholar] [CrossRef]
  36. Wang, H.; Cao, Y.; Gou, Y.; Wang, H.; Liang, Z.; Wu, Q.; Tan, J.; Liu, J.; Li, Z.; Cui, J.; et al. IGF2BP3 promotes glutamine metabolism of endometriosis by interacting with UCA1 to enhances the mRNA stability of GLS1. Mol. Med. 2024, 30, 64. [Google Scholar] [CrossRef] [PubMed]
  37. Hsiao, K.-Y.; Lin, S.-C.; Wu, M.-H.; Tsai, S.-J. Pathological functions of hypoxia in endometriosis. Front. Biosci. (Elite Ed.) 2015, 7, 309–321. [Google Scholar] [CrossRef]
  38. Li, W.N.; Wu, M.H.; Tsai, S.J. HYPOXIA AND REPRODUCTIVE HEALTH: The role of hypoxia in the development and progression of endometriosis. Reproduction 2021, 161, F19–F31. [Google Scholar] [CrossRef] [PubMed]
  39. Rytkonen, K.T.; Heinosalo, T.; Mahmoudian, M.; Ma, X.; Perheentupa, A.; Elo, L.L.; Poutanen, M.; Wagner, G.P. Transcriptomic responses to hypoxia in endometrial and decidual stromal cells. Reproduction 2020, 160, 39–51. [Google Scholar] [CrossRef]
  40. Lac, V.; Nazeran, T.M.; Tessier-Cloutier, B.; Aguirre-Hernandez, R.; Albert, A.; Lum, A.; Khattra, J.; Praetorius, T.; Mason, M.; Chiu, D.; et al. Oncogenic mutations in histologically normal endometrium: The new normal? J. Pathol. 2019, 249, 173–181. [Google Scholar] [CrossRef]
  41. Suda, K.; Nakaoka, H.; Yoshihara, K.; Ishiguro, T.; Tamura, R.; Mori, Y.; Yamawaki, K.; Adachi, S.; Takahashi, T.; Kase, H.; et al. Clonal Expansion and Diversification of Cancer-Associated Mutations in Endometriosis and Normal Endometrium. Cell Rep. 2018, 24, 1777–1789. [Google Scholar] [CrossRef]
  42. Pocate-Cheriet, K.; Santulli, P.; Kateb, F.; Bourdon, M.; Maignien, C.; Batteux, F.; Chouzenoux, S.; Patrat, C.; Wolf, J.P.; Bertho, G.; et al. The follicular fluid metabolome differs according to the endometriosis phenotype. Reprod. Biomed. Online 2020, 41, 1023–1037. [Google Scholar] [CrossRef]
  43. McKinnon, B.; Bertschi, D.; Wotzkow, C.; Bersinger, N.A.; Evers, J.; Mueller, M.D. Glucose transporter expression in eutopic endometrial tissue and ectopic endometriotic lesions. J. Mol. Endocrinol. 2014, 52, 169–179. [Google Scholar] [CrossRef] [PubMed]
  44. Kierans, S.J.; Taylor, C.T. Regulation of glycolysis by the hypoxia-inducible factor (HIF): Implications for cellular physiology. J. Physiol. 2021, 599, 23–37. [Google Scholar] [CrossRef] [PubMed]
  45. Peng, J.; Wang, X.; Ran, L.; Song, J.; Luo, R.; Wang, Y. Hypoxia-Inducible Factor 1alpha Regulates the Transforming Growth Factor beta1/SMAD Family Member 3 Pathway to Promote Breast Cancer Progression. J. Breast Cancer 2018, 21, 259–266. [Google Scholar] [CrossRef] [PubMed]
  46. Li, J.; Liu, L.; Fan, R. The PKM2/HIF-1alpha Axis is Involved in the Pathogenesis of Endometriosis via TGF-beta1 under Endometrial Polyps. Front. Biosci. (Landmark Ed.) 2024, 29, 417. [Google Scholar] [CrossRef]
  47. Sarsenova, M.; Boggavarapu, N.R.; Kask, K.; Modhukur, V.; Samuel, K.; Karro, H.; Gemzell-Danielsson, K.; Lalitkumar, P.G.L.; Salumets, A.; Peters, M.; et al. Hypoxic conditions affect transcriptome of endometrial stromal cells in endometriosis and promote TGFBI axis. Front. Endocrinol. 2024, 15, 1465393. [Google Scholar] [CrossRef]
  48. Skuli, N.; Majmundar, A.J.; Krock, B.L.; Mesquita, R.C.; Mathew, L.K.; Quinn, Z.L.; Runge, A.; Liu, L.; Kim, M.N.; Liang, J.; et al. Endothelial HIF-2alpha regulates murine pathological angiogenesis and revascularization processes. J. Clin. Investig. 2012, 122, 1427–1443. [Google Scholar] [CrossRef]
  49. Rankin, E.B.; Rha, J.; Unger, T.L.; Wu, C.H.; Shutt, H.P.; Johnson, R.S.; Simon, M.C.; Keith, B.; Haase, V.H. Hypoxia-inducible factor-2 regulates vascular tumorigenesis in mice. Oncogene 2008, 27, 5354–5358. [Google Scholar] [CrossRef]
  50. Iommarini, L.; Porcelli, A.M.; Gasparre, G.; Kurelac, I. Non-Canonical Mechanisms Regulating Hypoxia-Inducible Factor 1 Alpha in Cancer. Front. Oncol. 2017, 7, 286. [Google Scholar] [CrossRef]
  51. Ngo, C.; Chereau, C.; Nicco, C.; Weill, B.; Chapron, C.; Batteux, F. Reactive oxygen species controls endometriosis progression. Am. J. Pathol. 2009, 175, 225–234. [Google Scholar] [CrossRef]
  52. Dai, W.; Guo, R.; Na, X.; Jiang, S.; Liang, J.; Guo, C.; Fang, Y.; Na, Z.; Li, D. Hypoxia and the endometrium: An indispensable role for HIF-1α as therapeutic strategies. Redox Biol. 2024, 73, 103205. [Google Scholar] [CrossRef] [PubMed]
  53. Jung, Y.J.; Isaacs, J.S.; Lee, S.; Trepel, J.; Neckers, L. IL-1beta-mediated up-regulation of HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB J. 2003, 17, 2115–2117. [Google Scholar] [CrossRef] [PubMed]
  54. Liang, S.; Dong, S.; Liu, W.; Wang, M.; Tian, S.; Ai, Y.; Wang, H. Accumulated ROS Activates HIF-1alpha-Induced Glycolysis and Exerts a Protective Effect on Sensory Hair Cells Against Noise-Induced Damage. Front. Mol. Biosci. 2021, 8, 806650. [Google Scholar] [CrossRef]
  55. Lingappan, K. NF-kappaB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
  56. Gao, J.; Zhao, Y.; Li, T.; Gan, X.; Yu, H. The Role of PKM2 in the Regulation of Mitochondrial Function: Focus on Mitochondrial Metabolism, Oxidative Stress, Dynamic, and Apoptosis. PKM2 in Mitochondrial Function. Oxid. Med. Cell. Longev. 2022, 2022, 7702681. [Google Scholar] [CrossRef]
  57. Barra, F.; Ferro Desideri, L.; Ferrero, S. Inhibition of PI3K/AKT/mTOR pathway for the treatment of endometriosis. Br. J. Pharmacol. 2018, 175, 3626–3627. [Google Scholar] [CrossRef] [PubMed]
  58. Makker, A.; Goel, M.M.; Das, V.; Agarwal, A. PI3K-Akt-mTOR and MAPK signaling pathways in polycystic ovarian syndrome, uterine leiomyomas and endometriosis: An update. Gynecol. Endocrinol. 2012, 28, 175–181. [Google Scholar] [CrossRef]
  59. Assaf, L.; Eid, A.A.; Nassif, J. Role of AMPK/mTOR, mitochondria, and ROS in the pathogenesis of endometriosis. Life Sci. 2022, 306, 120805. [Google Scholar] [CrossRef]
  60. Zhang, J.; Kim, S.; Li, L.; Kemp, C.J.; Jiang, C.; Lu, J. Proteomic and transcriptomic profiling of Pten gene-knockout mouse model of prostate cancer. Prostate 2020, 80, 588–605. [Google Scholar] [CrossRef]
  61. Xi, Y.; Qi, Z.; Ma, J.; Chen, Y. PTEN loss activates a functional AKT/CXCR4 signaling axis to potentiate tumor growth and lung metastasis in human osteosarcoma cells. Clin. Exp. Metastasis 2020, 37, 173–185. [Google Scholar] [CrossRef]
  62. Dai, F.; Li, J.; Liu, Y. Phosphatase and tensin homolog deficiency induces M2 macrophage polarization by promoting glycolytic activity in endometrial stromal cells. Biol. Reprod. 2025, 112, 640–650. [Google Scholar] [CrossRef] [PubMed]
  63. Jiang, W.; He, T.; Liu, S.; Zheng, Y.; Xiang, L.; Pei, X.; Wang, Z.; Yang, H. The PIK3CA E542K and E545K mutations promote glycolysis and proliferation via induction of the beta-catenin/SIRT3 signaling pathway in cervical cancer. J. Hematol. Oncol. 2018, 11, 139. [Google Scholar] [CrossRef] [PubMed]
  64. Young, V.J.; Ahmad, S.F.; Brown, J.K.; Duncan, W.C.; Horne, A.W. ID2 mediates the transforming growth factor-beta1-induced Warburg-like effect seen in the peritoneum of women with endometriosis. Mol. Hum. Reprod. 2016, 22, 648–654. [Google Scholar] [CrossRef]
  65. Remels, A.; Gosker, H.; Verhees, K.; Langen, R.; Schols, A. TNF-α-induced NF-κB activation stimulates skeletal muscle glycolytic metabolism through activation of HIF-1α. Endocrinology 2015, 156, 1770–1781. [Google Scholar] [CrossRef] [PubMed]
  66. Camporeale, A.; Demaria, M.; Monteleone, E.; Giorgi, C.; Wieckowski, M.R.; Pinton, P.; Poli, V. STAT3 Activities and Energy Metabolism: Dangerous Liaisons. Cancers 2014, 6, 1579–1596. [Google Scholar] [CrossRef]
  67. Luckow Invitti, A.; Schor, E.; Martins Parreira, R.; Kopelman, A.; Kamergorodsky, G.; Gonçalves, G.A.; Batista Castello Girão, M.J. Inflammatory cytokine profile of co-cultivated primary cells from the endometrium of women with and without endometriosis. Mol. Med. Rep. 2018, 18, 1287–1296. [Google Scholar] [CrossRef]
  68. Taylor, D.J.; Faragher, E.B.; Evanson, J.M. Inflammatory cytokines stimulate glucose uptake and glycolysis but reduce glucose oxidation in human dermal fibroblasts in vitro. Circ. Shock 1992, 37, 105–110. [Google Scholar]
  69. Pak, H.K.; Nam, B.; Lee, Y.K.; Kim, Y.W.; Roh, J.; Son, J.; Chung, Y.S.; Choe, J.; Park, C.S. Human Plasmablast Migration Toward CXCL12 Requires Glucose Oxidation by Enhanced Pyruvate Dehydrogenase Activity via AKT. Front. Immunol. 2018, 9, 1742. [Google Scholar] [CrossRef]
  70. Luker, K.E.; Luker, G.D. The CXCL12/CXCR4/ACKR3 Signaling Axis Regulates PKM2 and Glycolysis. Cells 2022, 11, 1775. [Google Scholar] [CrossRef]
  71. de Azambuja Rodrigues, P.M.; Valente, R.H.; Brunoro, G.V.F.; Nakaya, H.T.I.; Araujo-Pereira, M.; Bozza, P.T.; Bozza, F.A.; Trugilho, M.R.O. Proteomics reveals disturbances in the immune response and energy metabolism of monocytes from patients with septic shock. Sci. Rep. 2021, 11, 15149. [Google Scholar] [CrossRef]
  72. Luo, Y.; Jiang, Q.; Zhu, Z.; Sattar, H.; Wu, J.; Huang, W.; Su, S.; Liang, Y.; Wang, P.; Meng, X. Phosphoproteomics and Proteomics Reveal Metabolism as a Key Node in LPS-Induced Acute Inflammation in RAW264.7. Inflammation 2020, 43, 1667–1679. [Google Scholar] [CrossRef] [PubMed]
  73. Sun, L.; Yang, X.; Yuan, Z.; Wang, H. Metabolic Reprogramming in Immune Response and Tissue Inflammation. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1990–2001. [Google Scholar] [CrossRef] [PubMed]
  74. Penariol, L.B.; Thomé, C.H.; Tozetti, P.A.; Paier, C.R.; Buono, F.O.; Peronni, K.C.; Orellana, M.D.; Covas, D.T.; Moraes, M.E.; Silva, W.A., Jr. What do the transcriptome and proteome of menstrual blood-derived mesenchymal stem cells tell us about endometriosis? Int. J. Mol. Sci. 2022, 23, 11515. [Google Scholar] [CrossRef] [PubMed]
  75. Kasvandik, S.; Samuel, K.; Peters, M.; Eimre, M.; Peet, N.; Roost, A.M.; Padrik, L.; Paju, K.; Peil, L.; Salumets, A. Deep Quantitative Proteomics Reveals Extensive Metabolic Reprogramming and Cancer-Like Changes of Ectopic Endometriotic Stromal Cells. J. Proteome Res. 2016, 15, 572–584. [Google Scholar] [CrossRef]
  76. Sapkota, Y.; Steinthorsdottir, V.; Morris, A.P.; Fassbender, A.; Rahmioglu, N.; De Vivo, I.; Buring, J.E.; Zhang, F.; Edwards, T.L.; Jones, S.; et al. Meta-analysis identifies five novel loci associated with endometriosis highlighting key genes involved in hormone metabolism. Nat. Commun. 2017, 8, 15539. [Google Scholar] [CrossRef]
  77. Ducreux, B.; Patrat, C.; Firmin, J.; Ferreux, L.; Chapron, C.; Marcellin, L.; Parpex, G.; Bourdon, M.; Vaiman, D.; Santulli, P.; et al. Systematic review on the DNA methylation role in endometriosis: Current evidence and perspectives. Clin. Epigenet. 2025, 17, 32. [Google Scholar] [CrossRef]
  78. Zubrzycka, A.; Zubrzycki, M.; Perdas, E.; Zubrzycka, M. Genetic, Epigenetic, and Steroidogenic Modulation Mechanisms in Endometriosis. J. Clin. Med. 2020, 9, 1309. [Google Scholar] [CrossRef]
  79. Adammek, M.; Greve, B.; Kassens, N.; Schneider, C.; Bruggemann, K.; Schuring, A.N.; Starzinski-Powitz, A.; Kiesel, L.; Gotte, M. MicroRNA miR-145 inhibits proliferation, invasiveness, and stem cell phenotype of an in vitro endometriosis model by targeting multiple cytoskeletal elements and pluripotency factors. Fertil. Steril. 2013, 99, 1346–1355.e1345. [Google Scholar] [CrossRef]
  80. Bjorkman, S.; Taylor, H.S. MicroRNAs in endometriosis: Biological function and emerging biomarker candidatesdagger. Biol. Reprod. 2019, 100, 1135–1146. [Google Scholar] [CrossRef]
  81. Okamoto, M.; Nasu, K.; Abe, W.; Aoyagi, Y.; Kawano, Y.; Kai, K.; Moriyama, M.; Narahara, H. Enhanced miR-210 expression promotes the pathogenesis of endometriosis through activation of signal transducer and activator of transcription 3. Hum. Reprod. 2015, 30, 632–641. [Google Scholar] [CrossRef]
  82. Huang, P.; Zhu, S.; Liang, X.; Zhang, Q.; Luo, X.; Liu, C.; Song, L. Regulatory Mechanisms of LncRNAs in Cancer Glycolysis: Facts and Perspectives. Cancer Manag. Res. 2021, 13, 5317–5336. [Google Scholar] [CrossRef] [PubMed]
  83. Malakar, P.; Stein, I.; Saragovi, A.; Winkler, R.; Stern-Ginossar, N.; Berger, M.; Pikarsky, E.; Karni, R. Long Noncoding RNA MALAT1 Regulates Cancer Glucose Metabolism by Enhancing mTOR-Mediated Translation of TCF7L2. Cancer Res. 2019, 79, 2480–2493. [Google Scholar] [CrossRef]
  84. Ji, X.; Sun, W.; Lv, C.; Huang, J.; Zhang, H. Circular RNAs Regulate Glucose Metabolism in Cancer Cells. Onco Targets Ther. 2021, 14, 4005–4021. [Google Scholar] [CrossRef]
  85. Li, L.; Sun, B.; Sun, Y. Identification of functional TF-miRNA-hub gene regulatory network associated with ovarian endometriosis. Front. Genet. 2022, 13, 998417. [Google Scholar] [CrossRef] [PubMed]
  86. Wen, X.; Zhang, J.; Xu, Z.; Li, M.; Dong, X.; Du, Y.; Xu, Z.; Yan, L. Highly expressed lncRNA H19 in endometriosis promotes aerobic glycolysis and histone lactylation. Reproduction 2024, 168, e240018. [Google Scholar] [CrossRef]
  87. Qi, X.; Zhang, Y.; Ji, H.; Wu, X.; Wang, F.; Xie, M.; Shu, L.; Jiang, S.; Mao, Y.; Cui, Y.; et al. Knockdown of prohibitin expression promotes glucose metabolism in eutopic endometrial stromal cells from women with endometriosis. Reprod. Biomed. Online 2014, 29, 761–770. [Google Scholar] [CrossRef] [PubMed]
  88. Mao, J.; Zhang, J.; Cai, L.; Cui, Y.; Liu, J.; Mao, Y. Elevated prohibitin 1 expression mitigates glucose metabolism defects in granulosa cells of infertile patients with endometriosis. Mol. Hum. Reprod. 2022, 28, gaac018. [Google Scholar] [CrossRef]
  89. Signorile, A.; Sgaramella, G.; Bellomo, F.; De Rasmo, D. Prohibitins: A Critical Role in Mitochondrial Functions and Implication in Diseases. Cells 2019, 8, 71. [Google Scholar] [CrossRef]
  90. Zong, Y.; Li, H.; Liao, P.; Chen, L.; Pan, Y.; Zheng, Y.; Zhang, C.; Liu, D.; Zheng, M.; Gao, J. Mitochondrial dysfunction: Mechanisms and advances in therapy. Signal Transduct. Target. Ther. 2024, 9, 124. [Google Scholar] [CrossRef]
  91. Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef]
  92. Kuhl, I.; Miranda, M.; Atanassov, I.; Kuznetsova, I.; Hinze, Y.; Mourier, A.; Filipovska, A.; Larsson, N.G. Transcriptomic and proteomic landscape of mitochondrial dysfunction reveals secondary coenzyme Q deficiency in mammals. eLife 2017, 6, e30952. [Google Scholar] [CrossRef] [PubMed]
  93. Kobayashi, H.; Matsubara, S.; Yoshimoto, C.; Shigetomi, H.; Imanaka, S. The role of mitochondrial dynamics in the pathophysiology of endometriosis. J. Obstet. Gynaecol. Res. 2023, 49, 2783–2791. [Google Scholar] [CrossRef]
  94. Zhan, L.; Wang, W.; Zhang, Y.; Song, E.; Fan, Y.; Wei, B. Hypoxia-inducible factor-1alpha: A promising therapeutic target in endometriosis. Biochimie 2016, 123, 130–137. [Google Scholar] [CrossRef]
  95. Hirschhaeuser, F.; Sattler, U.G.; Mueller-Klieser, W. Lactate: A metabolic key player in cancer. Cancer Res. 2011, 71, 6921–6925. [Google Scholar] [CrossRef]
  96. Marianna, S.; Alessia, P.; Susan, C.; Francesca, C.; Angela, S.; Francesca, C.; Antonella, N.; Patrizia, I.; Nicola, C.; Emilio, C. Metabolomic profiling and biochemical evaluation of the follicular fluid of endometriosis patients. Mol. Biosyst. 2017, 13, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  97. Ke, J.; Ye, J.; Li, M.; Zhu, Z. The Role of Matrix Metalloproteinases in Endometriosis: A Potential Target. Biomolecules 2021, 11, 1739. [Google Scholar] [CrossRef]
  98. Niland, S.; Riscanevo, A.X.; Eble, J.A. Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int. J. Mol. Sci. 2021, 23, 146. [Google Scholar] [CrossRef]
  99. Lee, H.C.; Lin, S.C.; Wu, M.H.; Tsai, S.J. Induction of Pyruvate Dehydrogenase Kinase 1 by Hypoxia Alters Cellular Metabolism and Inhibits Apoptosis in Endometriotic Stromal Cells. Reprod. Sci. 2019, 26, 734–744. [Google Scholar] [CrossRef] [PubMed]
  100. Horne, A.W.; Ahmad, S.F.; Carter, R.; Simitsidellis, I.; Greaves, E.; Hogg, C.; Morton, N.M.; Saunders, P.T.K. Repurposing dichloroacetate for the treatment of women with endometriosis. Proc. Natl. Acad. Sci. USA 2019, 116, 25389–25391. [Google Scholar] [CrossRef]
  101. Leow, H.W.; Koscielniak, M.; Williams, L.; Saunders, P.T.K.; Daniels, J.; Doust, A.M.; Jones, M.C.; Ferguson, G.D.; Bagger, Y.; Horne, A.W.; et al. Dichloroacetate as a possible treatment for endometriosis-associated pain: A single-arm open-label exploratory clinical trial (EPiC). Pilot Feasibility Stud. 2021, 7, 67. [Google Scholar] [CrossRef]
  102. Kim, B.S.; Chung, T.W.; Choi, H.J.; Bae, S.J.; Cho, H.R.; Lee, S.O.; Choi, J.H.; Joo, J.K.; Ha, K.T. Caesalpinia sappan induces apoptotic cell death in ectopic endometrial 12Z cells through suppressing pyruvate dehydrogenase kinase 1 expression. Exp. Ther. Med. 2021, 21, 357. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, H.; Liang, Z.; Gou, Y.; Li, Z.; Cao, Y.; Jiao, N.; Tan, J.; Yu, Y.; Zhang, Z. FTO-dependent N(6)-Methyladenosine regulates the progression of endometriosis via the ATG5/PKM2 Axis. Cell Signal. 2022, 98, 110406. [Google Scholar] [CrossRef]
  104. Zheng, J.; Dai, Y.; Lin, X.; Huang, Q.; Shi, L.; Jin, X.; Liu, N.; Zhou, F.; Zhang, S. Hypoxia-induced lactate dehydrogenase A protects cells from apoptosis in endometriosis. Mol. Med. Rep. 2021, 24, 637. [Google Scholar] [CrossRef]
  105. Cho, M.K.; Jin, L.; Han, J.H.; Jin, J.S.; Cheon, S.Y.; Shin, S.; Bae, S.J.; Park, J.K.; Ha, K.T. Water-Extracted Prunella vulgaris Alleviates Endometriosis by Reducing Aerobic Glycolysis. Front. Pharmacol. 2022, 13, 872810. [Google Scholar] [CrossRef]
  106. Han, J.H.; Lee, E.J.; Park, W.; Ha, K.T.; Chung, H.S. Natural compounds as lactate dehydrogenase inhibitors: Potential therapeutics for lactate dehydrogenase inhibitors-related diseases. Front. Pharmacol. 2023, 14, 1275000. [Google Scholar] [CrossRef] [PubMed]
  107. Yao, Q.; Jing, G.; Zhang, X.; Li, M.; Yao, Q.; Wang, L. Cinnamic acid inhibits cell viability, invasion, and glycolysis in primary endometrial stromal cells by suppressing NF-kappaB-induced transcription of PKM2. Biosci. Rep. 2021, 9, BSR20211828. [Google Scholar] [CrossRef]
  108. Wang, M.; Fan, R.; Jiang, J.; Sun, F.; Sun, Y.; Wang, Q.; Jiang, A.; Yu, Z.; Yang, T. PIM2 Promotes the Development of Ovarian Endometriosis by Enhancing Glycolysis and Fibrosis. Reprod. Sci. 2023, 30, 2692–2702. [Google Scholar] [CrossRef] [PubMed]
  109. Lu, C.; Qiao, P.; Fu, R.; Wang, Y.; Lu, J.; Ling, X.; Liu, L.; Sun, Y.; Ren, C.; Yu, Z. Phosphorylation of PFKFB4 by PIM2 promotes anaerobic glycolysis and cell proliferation in endometriosis. Cell Death Dis. 2022, 13, 790. [Google Scholar] [CrossRef]
  110. Lu, J.; Wang, X.; Shi, X.; Jiang, J.; Liu, L.; Liu, L.; Ren, C.; Lu, C.; Yu, Z. PAK5-mediated PKM2 phosphorylation is critical for anaerobic glycolysis in endometriosis. Front. Med. 2024, 18, 1054–1067. [Google Scholar] [CrossRef]
  111. Zhou, J.; Ding, Z.M.; Hardiman, P.J. Understanding the Role of Gui-Zhi-Fu-Ling-Capsules (Chinese Medicine) for Treatment of Endometriosis in the Rat Model: Using NMR Based Metabolomics. Evid. Based Complement. Altern. Med. 2018, 2018, 9864963. [Google Scholar] [CrossRef]
  112. Bahrami, A.; Ayen, E.; Razi, M.; Behfar, M. Effects of atorvastatin and resveratrol against the experimental endometriosis; evidence for glucose and monocarboxylate transporters, neoangiogenesis. Life Sci. 2021, 272, 119230. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, Y.; Xiu, J.; Yang, T.; Ren, C.; Yu, Z. HSF1 promotes endometriosis development and glycolysis by up-regulating PFKFB3 expression. Reprod. Biol. Endocrinol. 2021, 19, 86. [Google Scholar] [CrossRef]
  114. Ling, X.; Lu, J.; Wang, X.; Liu, L.; Liu, L.; Wang, Y.; Sun, Y.; Ren, C.; Lu, C.; Yu, Z. Ovarian tumorB1-mediated heat shock transcription factor 1 deubiquitination is critical for glycolysis and development of endometriosis. iScience 2022, 25, 105363. [Google Scholar] [CrossRef] [PubMed]
  115. Sun, Y.; Wang, Q.; Wang, M.; Sun, F.; Qiao, P.; Jiang, A.; Ren, C.; Yu, Z.; Yang, T. CHIP induces ubiquitination and degradation of HMGB1 to regulate glycolysis in ovarian endometriosis. Cell Mol. Life Sci. 2022, 80, 13. [Google Scholar] [CrossRef] [PubMed]
  116. Gou, Y.; Wang, H.; Wang, T.; Wang, H.; Wang, B.; Jiao, N.; Yu, Y.; Cao, Y.; Wang, H.; Zhang, Z. Ectopic endometriotic stromal cells-derived lactate induces M2 macrophage polarization via Mettl3/Trib1/ERK/STAT3 signalling pathway in endometriosis. Immunology 2023, 168, 389–402. [Google Scholar] [CrossRef]
  117. Ling, X.; Liu, L.; Jiang, A.; Shi, X.; Liu, L.; Wang, X.; Lu, C.; Ren, C.; Yu, Z. PFKFB3 promotes endometriosis cell proliferation via enhancing the protein stability of beta-catenin. Mol. Cell. Endocrinol. 2024, 579, 112083. [Google Scholar] [CrossRef]
  118. Sun, Y.; Zhang, S.; Zhang, X.; Li, G.; Sun, F.; Wang, M.; Ren, C.; Jiang, A.; Yang, T. AURKA Enhances the Glycolysis and Development of Ovarian Endometriosis Through ERbeta. Endocrinology 2024, 165, bqae018. [Google Scholar] [CrossRef]
  119. Huang, Z.X.; Lin, D.C.; Zhang, H.Y.; Yang, M.J.; Chen, J.H.; Ding, X.Y.; Dai, S.J.; Hong, Y.H.; Liang, G.S.; Li, Q.Y.; et al. The dysfunction of CD8(+) T cells triggered by endometriotic stromal cells promotes the immune survival of endometriosis. Immunology 2024, 172, 469–485. [Google Scholar] [CrossRef]
  120. Gao, X.; Shao, W.; Wang, J.; Gao, H.; Zhang, X.; Xia, C.; Li, M.; Liu, S. Integrin beta3 enhances glycolysis and increases lactate production in endometriosis. J. Reprod. Immunol. 2024, 165, 104312. [Google Scholar] [CrossRef]
  121. Liu, Y.; Xu, R.; Gu, H.; Zhang, E.; Qu, J.; Cao, W.; Huang, X.; Yan, H.; He, J.; Cai, Z. Metabolic reprogramming in macrophage responses. Biomark. Res. 2021, 9, 1. [Google Scholar] [CrossRef]
  122. Yee, J.L.; Huang, C.Y.; Yu, Y.C.; Huang, S.J. Potential Mechanisms of Guizhi Fuling Wan in Treating Endometriosis: An Analysis Based on TCMSP and DisGeNET Databases. J. Ethnopharmacol. 2024, 329, 118190. [Google Scholar] [CrossRef]
  123. Medina, M.G.; Lebovic, D.I. Endometriosis-associated nerve fibers and pain. Acta Obstet. Gynecol. Scand. 2009, 88, 968–975. [Google Scholar] [CrossRef] [PubMed]
  124. Mohammed Rasheed, H.A.; Hamid, P. Inflammation to Infertility: Panoramic View on Endometriosis. Cureus 2020, 12, e11516. [Google Scholar] [CrossRef]
  125. Fan, W.; Yuan, Z.; Li, M.; Zhang, Y.; Nan, F. Decreased oocyte quality in patients with endometriosis is closely related to abnormal granulosa cells. Front. Endocrinol. 2023, 14, 1226687. [Google Scholar] [CrossRef] [PubMed]
  126. Kobayashi, H.; Kimura, M.; Maruyama, S.; Nagayasu, M.; Imanaka, S. Revisiting estrogen-dependent signaling pathways in endometriosis: Potential targets for non-hormonal therapeutics. Eur. J. Obstet. Gynecol. Reprod. Biol. 2021, 258, 103–110. [Google Scholar] [CrossRef] [PubMed]
  127. Scutiero, G.; Iannone, P.; Bernardi, G.; Bonaccorsi, G.; Spadaro, S.; Volta, C.A.; Greco, P.; Nappi, L. Oxidative Stress and Endometriosis: A Systematic Review of the Literature. Oxid. Med. Cell. Longev. 2017, 2017, 7265238. [Google Scholar] [CrossRef]
  128. Song, S.Y.; Jung, Y.W.; Shin, W.; Park, M.; Lee, G.W.; Jeong, S.; An, S.; Kim, K.; Ko, Y.B.; Lee, K.H.; et al. Endometriosis-Related Chronic Pelvic Pain. Biomedicines 2023, 11, 2868. [Google Scholar] [CrossRef]
  129. Kalaitzopoulos, D.R.; Samartzis, N.; Kolovos, G.N.; Mareti, E.; Samartzis, E.P.; Eberhard, M.; Dinas, K.; Daniilidis, A. Treatment of endometriosis: A review with comparison of 8 guidelines. BMC Womens Health 2021, 21, 397. [Google Scholar] [CrossRef]
  130. Capezzuoli, T.; Rossi, M.; La Torre, F.; Vannuccini, S.; Petraglia, F. Hormonal drugs for the treatment of endometriosis. Curr. Opin. Pharmacol. 2022, 67, 102311. [Google Scholar] [CrossRef]
Figure 1. Molecular and metabolic pathways underlying the Warburg’s effect in cancer. Cancer cells preferentially utilize aerobic glycolysis over mitochondrial oxidative phosphorylation even in the presence of oxygen, a phenomenon known as the Warburg effect. This metabolic reprogramming is regulated by several oncogenic and tumor-suppressive pathways. HIF-1α, stabilized under hypoxic or oncogenic conditions, promotes the expression of GLUT1 and GLUT3 and PDK1 and PDK3, which inhibit PDH, thereby blocking the entry of pyruvate into the TCA cycle. The PI3K/AKT/mTOR pathway enhances glycolysis by upregulating GLUT1 and HK2 while supporting cell proliferation and survival. Conversely, the tumor suppressor p53 inhibits glycolysis via TIGAR and enhances oxidative phosphorylation through SCO2. Loss of p53 in tumors removes these inhibitory mechanisms, allowing glycolysis to proceed unchecked. Cancer cells also exhibit metabolic plasticity, adapting between glycolysis and oxidative phosphorylation, depending on the environmental conditions, and frequently increase glutamine metabolism to fuel the TCA cycle. These adaptations not only support biosynthesis and rapid growth but also promote invasion, metastasis, and immune evasion, contributing to therapy resistance. The red asterisks indicate the molecular targets of therapeutic drugs.
Figure 1. Molecular and metabolic pathways underlying the Warburg’s effect in cancer. Cancer cells preferentially utilize aerobic glycolysis over mitochondrial oxidative phosphorylation even in the presence of oxygen, a phenomenon known as the Warburg effect. This metabolic reprogramming is regulated by several oncogenic and tumor-suppressive pathways. HIF-1α, stabilized under hypoxic or oncogenic conditions, promotes the expression of GLUT1 and GLUT3 and PDK1 and PDK3, which inhibit PDH, thereby blocking the entry of pyruvate into the TCA cycle. The PI3K/AKT/mTOR pathway enhances glycolysis by upregulating GLUT1 and HK2 while supporting cell proliferation and survival. Conversely, the tumor suppressor p53 inhibits glycolysis via TIGAR and enhances oxidative phosphorylation through SCO2. Loss of p53 in tumors removes these inhibitory mechanisms, allowing glycolysis to proceed unchecked. Cancer cells also exhibit metabolic plasticity, adapting between glycolysis and oxidative phosphorylation, depending on the environmental conditions, and frequently increase glutamine metabolism to fuel the TCA cycle. These adaptations not only support biosynthesis and rapid growth but also promote invasion, metastasis, and immune evasion, contributing to therapy resistance. The red asterisks indicate the molecular targets of therapeutic drugs.
Pharmaceuticals 18 00813 g001
Figure 2. Metabolic reprogramming in endometriosis: A shift toward glycolysis and mitochondrial dysfunction. Ectopic endometriotic cells exhibit distinct metabolic alterations compared to normal endometrial cells, including a shift toward aerobic glycolysis (Warburg effect), increased fatty acid oxidation, and mitochondrial dysfunction. Hypoxia in the peritoneal cavity activates HIF-1α, which enhances glycolytic gene expression and suppresses oxidative phosphorylation. These changes result in elevated lactate production, promoting an acidic and inflammatory microenvironment that facilitates immune evasion and lesion survival. Enhanced fatty acid oxidation provides alternative energy under stress, while glutamine metabolism supports NADPH production and biosynthesis. Mitochondrial dysfunction, characterized by impaired electron transport, increased ROS, and mtDNA mutations, further drives disease progression.
Figure 2. Metabolic reprogramming in endometriosis: A shift toward glycolysis and mitochondrial dysfunction. Ectopic endometriotic cells exhibit distinct metabolic alterations compared to normal endometrial cells, including a shift toward aerobic glycolysis (Warburg effect), increased fatty acid oxidation, and mitochondrial dysfunction. Hypoxia in the peritoneal cavity activates HIF-1α, which enhances glycolytic gene expression and suppresses oxidative phosphorylation. These changes result in elevated lactate production, promoting an acidic and inflammatory microenvironment that facilitates immune evasion and lesion survival. Enhanced fatty acid oxidation provides alternative energy under stress, while glutamine metabolism supports NADPH production and biosynthesis. Mitochondrial dysfunction, characterized by impaired electron transport, increased ROS, and mtDNA mutations, further drives disease progression.
Pharmaceuticals 18 00813 g002
Figure 3. Glycolysis-related metabolic changes, driven by genetic, epigenetic, and mitochondrial dysfunction, underlie endometriosis pathogenesis. Endometriotic cells undergo extensive metabolic reprogramming driven by hypoxia, inflammatory cytokines, genetic/epigenetic alterations, and mitochondrial dysfunction. Hypoxia in the peritoneal environment stabilizes HIF-1α and HIF-2α, enhancing glycolysis by upregulating GLUT1; GLUT4; glycolytic enzymes (HK2, PFK, and LDHA); and PDK1 while also promoting angiogenesis via VEGF. The PI3K/AKT/mTOR signaling cascade, often activated by PTEN loss or PIK3CA mutation, further boosts HIF-1α expression and glycolytic metabolism. Chronic inflammation involving TGF-β1, IL-6, IL-1β, and TNF-α activates NF-κB and STAT3 pathways, reinforcing the glycolytic phenotype and suppressing oxidative phosphorylation. Additionally, chemokines such as CXCL12/CXCR4 contribute to metabolic and migratory changes. Genetic mutations and epigenetic modifications (e.g., miRNAs, lncRNAs, and DNA methylation) influence the expression of key metabolic regulators. Mitochondrial dysfunction—evidenced by reduced oxidative phosphorylation enzyme expression, PHB loss, and excess ROS—contributes to HIF-1α stabilization, inflammation, and enhanced proliferation. These metabolic changes promote angiogenesis and ECM remodeling through increased lactate production, VEGF expression, and MMPs, ultimately driving lesion persistence and invasiveness.
Figure 3. Glycolysis-related metabolic changes, driven by genetic, epigenetic, and mitochondrial dysfunction, underlie endometriosis pathogenesis. Endometriotic cells undergo extensive metabolic reprogramming driven by hypoxia, inflammatory cytokines, genetic/epigenetic alterations, and mitochondrial dysfunction. Hypoxia in the peritoneal environment stabilizes HIF-1α and HIF-2α, enhancing glycolysis by upregulating GLUT1; GLUT4; glycolytic enzymes (HK2, PFK, and LDHA); and PDK1 while also promoting angiogenesis via VEGF. The PI3K/AKT/mTOR signaling cascade, often activated by PTEN loss or PIK3CA mutation, further boosts HIF-1α expression and glycolytic metabolism. Chronic inflammation involving TGF-β1, IL-6, IL-1β, and TNF-α activates NF-κB and STAT3 pathways, reinforcing the glycolytic phenotype and suppressing oxidative phosphorylation. Additionally, chemokines such as CXCL12/CXCR4 contribute to metabolic and migratory changes. Genetic mutations and epigenetic modifications (e.g., miRNAs, lncRNAs, and DNA methylation) influence the expression of key metabolic regulators. Mitochondrial dysfunction—evidenced by reduced oxidative phosphorylation enzyme expression, PHB loss, and excess ROS—contributes to HIF-1α stabilization, inflammation, and enhanced proliferation. These metabolic changes promote angiogenesis and ECM remodeling through increased lactate production, VEGF expression, and MMPs, ultimately driving lesion persistence and invasiveness.
Pharmaceuticals 18 00813 g003
Figure 4. Overview of the molecular targets and glycolysis inhibitors under investigation for endometriosis therapy. Multiple molecular targets and candidate compounds have been investigated to inhibit aerobic glycolysis in endometriotic cells. PDK1 and PDK3 are the most studied targets, with DCA being the leading compound shown to restore mitochondrial function, reduce lactate production, and alleviate pain. Natural extracts such as Caesalpinia sappan and Prunella vulgaris inhibit PDKs and LDHA, reducing glycolysis and promoting apoptosis. Other notable targets include PKM2, inhibited by succinic acid and PIM2 inhibitors (e.g., SMI-4a), and PAK5, which inhibition destabilizes PKM2 and suppresses glycolysis. PFKFB3 is targeted by PFK-015 and regulated by HSF1, which is inhibited by KRIBB11. The inhibition of HK2 with 2-DG also suppresses glycolysis and macrophage infiltration. Additional compounds like alisertib (AURKA inhibitor), YL-109 (CHIP agonist), and natural agents such as resveratrol and Gui-Zhi-Fu-Ling capsules show multi-targeted effects, including anti-glycolytic, anti-fibrotic, and anti-angiogenic actions.
Figure 4. Overview of the molecular targets and glycolysis inhibitors under investigation for endometriosis therapy. Multiple molecular targets and candidate compounds have been investigated to inhibit aerobic glycolysis in endometriotic cells. PDK1 and PDK3 are the most studied targets, with DCA being the leading compound shown to restore mitochondrial function, reduce lactate production, and alleviate pain. Natural extracts such as Caesalpinia sappan and Prunella vulgaris inhibit PDKs and LDHA, reducing glycolysis and promoting apoptosis. Other notable targets include PKM2, inhibited by succinic acid and PIM2 inhibitors (e.g., SMI-4a), and PAK5, which inhibition destabilizes PKM2 and suppresses glycolysis. PFKFB3 is targeted by PFK-015 and regulated by HSF1, which is inhibited by KRIBB11. The inhibition of HK2 with 2-DG also suppresses glycolysis and macrophage infiltration. Additional compounds like alisertib (AURKA inhibitor), YL-109 (CHIP agonist), and natural agents such as resveratrol and Gui-Zhi-Fu-Ling capsules show multi-targeted effects, including anti-glycolytic, anti-fibrotic, and anti-angiogenic actions.
Pharmaceuticals 18 00813 g004
Figure 5. Role of metabolic reprogramming in endometriosis pathophysiology and therapeutic strategies. The shift to glycolysis in endometriotic lesions increases lactate production, creating an acidic microenvironment that promotes angiogenesis, tissue invasion, and immune evasion. These changes lead to chronic inflammation, extracellular matrix remodeling, and increased nerve fiber density, contributing to pain and symptom severity. Metabolic alterations also impair fertility by affecting the oocyte quality and endometrial receptivity. While hormonal therapies target estrogen-driven proliferation, combining them with metabolic inhibitors like mTOR and glycolysis blockers may improve treatment outcomes. Anti-inflammatory and mitochondrial therapies could further reduce recurrence. However, most of the evidence is preclinical, underscoring the need for more clinical studies.
Figure 5. Role of metabolic reprogramming in endometriosis pathophysiology and therapeutic strategies. The shift to glycolysis in endometriotic lesions increases lactate production, creating an acidic microenvironment that promotes angiogenesis, tissue invasion, and immune evasion. These changes lead to chronic inflammation, extracellular matrix remodeling, and increased nerve fiber density, contributing to pain and symptom severity. Metabolic alterations also impair fertility by affecting the oocyte quality and endometrial receptivity. While hormonal therapies target estrogen-driven proliferation, combining them with metabolic inhibitors like mTOR and glycolysis blockers may improve treatment outcomes. Anti-inflammatory and mitochondrial therapies could further reduce recurrence. However, most of the evidence is preclinical, underscoring the need for more clinical studies.
Pharmaceuticals 18 00813 g005
Table 1. Summary of the articles retrieved from PubMed using the terms “glycolysis” and “endometriosis”.
Table 1. Summary of the articles retrieved from PubMed using the terms “glycolysis” and “endometriosis”.
First AuthorKey MechanismMolecular TargetDrugExperimentsPublication YearRef
McKinnon BAltered glucose metabolism is mediated by glucose transporterGLUT4-In vitro, In vivo2014[43]
Young VJTGF-β induces a Warburg-like effectHIF1A, PDK1, LDHA, GLUT1-In vitro, Human sample2014[10]
Qi XEnhanced glycolysis is due to prohibitin (PHB) downregulationPHB-In vitro, Human sample2014[87]
Kasvandik SWarburg effect leads to enhanced invasiveness, adhesion, and immune evasion.--Human sample, In silico2016[75]
Young VJTGF-β1-induced Warburg effect via suppression of ID2, which in turn upregulates HIF-1α expression.ID2-In vitro, Human sample2016[64]
Zhou JRegulation of glycolysis and gluconeogenesis leads to metabolic changes and modulation of TGF-β1, GLUT-4, and VEGF expressionTGFB1, GLUT4, VEGFGui-Zhi-Fu-Ling CapsulesIn vivo, In silico2018[111]
Lee HCHypoxia upregulates PDK1 to enhance glycolysisPDK1DichloroacetateIn vitro2019[99]
Horne AWTGF-β1 drives increased glycolysis, reduced mitochondrial respiration, and enhanced lactate production.PDK1DichloroacetateIn vitro, In vivo, Human sample2019[100]
Rytkönen KTHypoxia-driven transcription factors Jun/Fos and CEBP, alters glycolysis, epithelial-mesenchymal transition, and inflammatory pathways.Jun, Fos, CEBP-In vitro, Human sample2020[39]
Pocate-Cheriet KMetabolic reprogramming in follicular fluid is characterized by altered glycolysis, beta-oxidation, and mitochondrial dysfunction.--Human sample, In silico2020[42]
Bahrami AInhibition of glycolysis and neovascularization suppresses endometriosis development.GLUT1, GLUT3, MCT1, MCT4Atorvastatin and ResveratrolIn vivo2021[112]
Leow HWMetabolic reprogramming correction reduces endometriosis-associated pain.PDK1DichloroacetateHuman clinical trials2021[101]
Kim BSInhibition of aerobic glycolysis and induction of ROS-mitochondria-mediated apoptosis.PDK1, PDK3Caesalpinia sappanIn vitro2021[102]
Wang YHSF1 promotes glycolysis via upregulating PFKFB3 facilitates endometriosis progression.HSF1, PFKFB3KRIBB11 (HSF1 inhibitor)in vitro, In vivo2021[113]
Zheng JLDHA promotes glycolysis and inhibits mitochondrial function.LDHAshLDHAIn vitro, Human sample2021[104]
Yao QPKM2 inhibition and may serve as a potential treatment for endometriosis.PKM2Cinnamic acidIn vitro, Human sample2021[107]
Cho MKInhibition of aerobic glycolysis prevents lesion growth and metabolic adaptation.LDHA, PDK1, PDK3Prunella vulgarisIn vitro, In vivo2022[105]
Hou SDownregulation of HK2 reduces migration, invasion, and proliferation of endometrial stromal cells.HK2, STAT1-In vitro2022[11]
Mao JUpregulation of PHB1 enhances glucose metabolism, ATP synthesis, and ROS production.PHB1-In vitro, Human sample2022[88]
Wang HFTO regulates ATG5 expression through m6A methylation and in turn, suppresses the expression of PKM2.FTO, ATG5, PKM2-In vitro, Human sample2022[103]
Lu CThe PIM2-PFKFB4 axis drives glycolysis and cell growth, contributing to EM progression.PIM2, PFKFB4-In vitro, In vivo, Human sample2022[109]
Li LThree key feedback loops were discovered in the TF-miRNA-hub gene networkMYC, YY1, HIF1A, LDHA, RELA, miR-34a-5p, miR-155-5p, miR-93-5p-Human sample, In silico2022[85]
Ling XOTUB1 increase the HSF1 stability and enhances glycolysis, EMT, and progression of endometriosis.OTUB1, HSF1-In vitro, In vivo2022[114]
Sun YCHIP activation reduces HMGB1 expression, limiting the energy supply for endometriotic cell growth.CHIP, HMGB1YL-109 (CHIP agonist)In vitro, In vivo, Human sample2022[115]
Gou YGlycolysis-driven lactate accumulation promotes M2 macrophage polarization, enhancing endometriotic lesion invasion via the Mettl3/Trib1/ERK-STAT3 pathway.Mettl3, Trib1, ERK, STAT32-Deoxy-D-glucose (2-DG)In vitro, In vivo, Human sample2023[116]
Chen QGlycolysis-related genes influence immune cell infiltration, contributing to endometriosis progression.CHPF, CITED2, GPC3, PDK3, ADH6-Human sample, In silico2023[31]
Wang MPIM2 enhances glycolysis and fibrosis in endometriotic cells by upregulating PKM2, promoting endometriosis progression.PIM2, PKM2, HK2, SMH, Desmin, α-SMASMI-4a (PIM2 inhibitor)In vitro, In vivo, Human sample2023[108]
Ling XPFKFB3 enhances glycolysis, proliferation, migration, and invasion of endometriosis cells by stabilizing β-catenin, promoting epithelial-mesenchymal transition (EMT).PFKFB3, β-cateninPFK-015 (PFKFB3 inhibitor)In vitro, In vivo, Human sample2024[117]
Sun YURKA promotes proliferation, migration, invasion, and glycolysis in ovarian endometriosis by upregulating ERβ.AURKA, ERβAlisertib (AURKA inhibitor)In vitro, In vivo, Human sample2024[118]
Huang ZXEndometriosis involves dysfunctional CD8+ T cells, where glycolysis and the STAT1/PDCD1 pathway reduce T-cell cytotoxicity and promote lesion growth.CDK1, CCNB1, STAT1, PDCD1-In vitro, In vivo, Human sample, In silico2024[119]
Guo Sincreased PDK3 and GPC3 levels and decreased ADH6 in EMT patients, which correlated with lower oocyte quality and pregnancy rates.PDK3, GPC3, ADH6-Human sample2024[32]
Toniyan KAendometriomas and pelvic peritoneum lesions, oxygen absorption is significantly reduced, and there is a shift towards succinate utilization, suggesting a Warburg effect with increased glycolysis.HK2, PK-Human sample2024[33]
Khashchenko EPglycolysis reprogramming, mitochondrial biogenesis, and apoptosis suppression drive peritoneal endometriosisMCT2, PDK1, GLUT1, OPA1, DRP1, Beclin1, Bnip3, ERβ-Human sample2024[34]
Wen XH19 expression promotes abnormal glucose metabolism and histone lactylation, driving endometriosis progression.H19-In vitro, In vivo, Human sample2024[86]
Gao XMacrophage-induced ITGB3 upregulation promotes glycolysis and enhances proliferation, migration, and invasion in endometriosis.ITGB3-In vitro, In vivo, In silico2024[120]
Sarsenova MMetabolic reprogramming, including glutathione metabolism, oxidative phosphorylation, and glycolysis, occurs in response to AMPK signaling and HIF-1 signaling perivascular cells of endometriotic lesions.AMPK, HIF1-Human sample, In silico2024[30]
Lu JPAK5 is upregulated in endometriosis, promoting glycolysis by stabilizing PKM2.PAK5, PKM2GNE 2861 (PAK5 inhibitor)In vitro, In vivo, Human sample2024[110]
Li JPKM2/HIF-1α axis regulates glycolysis and plays a key role in endometriosis pathogenesis.TGFB1, HIF1A, PKM2-In vitro, Human sample2024[46]
Sarsenova MHypoxia-induced TGFBI is involved in fibrosis and angiogenesis and is more highly expressed in ectopic endometrial tissue during the secretory phase.TGFB1-In vitro, Human sample2024[47]
Dai FPTEN deficiency in ectopic endometrial stromal cells (EESCs) promotes glycolytic activity, which enhances M2 macrophage polarization and increases CCL2 production.PTEN, CCL2-In vitro2025[62]
Table 2. Comparative analysis of the candidate therapeutic agents targeting “glycolysis” and “endometriosis”: mechanisms, development stages, efficacy, and safety.
Table 2. Comparative analysis of the candidate therapeutic agents targeting “glycolysis” and “endometriosis”: mechanisms, development stages, efficacy, and safety.
Drug NameMechanismDevelopment StageEfficacySafety
Gui-Zhi-Fu-Ling CapsulesPromotes blood circulation and removes blood stasisApproved for clinical use (in China)Anti-inflammatory effects, alleviates abdominal pain, regulates menstruationGenerally safe; lacks standardized clinical data
Dichloroacetate (DCA)Inhibits pyruvate dehydrogenase kinase (PDK) to normalize glucose metabolismPreclinical/Phase 2Reduces peritoneal lactate levels and lesion size (animal models)Potential neurotoxicity with prolonged use
Atorvastatin + ResveratrolInhibits GLUT-1/3 and MCT-1/4 to suppress glycolysis and angiogenesisPreclinical40% reduction in lesion size and angiogenesis densityMuscle toxicity risk (statin-related)
Caesalpinia sappan ExtractPDK1 inhibition induces mitochondrial ROS and apoptosisPreclinical>50% apoptosis in 12Z cellsLimited human toxicity data
KRIBB11HSF1 inhibition reduces PFKFB3 and glycolysisPreclinical60% lesion weight reduction in animal modelsPotential heat shock protein dysregulation
shLDHALDHA gene silencing to inhibit lactate productionEarly research stage70% inhibition of 12Z cell migrationUnconfirmed long-term safety of gene therapy
Cinnamic AcidSuppresses NF-κB/PKM2 signaling to reduce invasivenessPreclinical55% reduction in stromal cell invasionMucosal irritation at high doses
Prunella vulgaris ExtractLDHA/PDK1 inhibition disrupts mitochondrial functionPreclinical40% decrease in oxygen consumption rate in 12Z cellsTraditional use history but lacking modern toxicity data
YL-109CHIP activation promotes HMGB1 ubiquitination and degradationPreclinical65% lesion size reduction in animal modelsUnclear immune system impacts
2-Deoxy-D-glucose (2-DG)Glycolysis blockade via glucose analogPreclinical75% disease progression inhibition in miceHypoglycemia risk
SMI-4aPIM2 inhibition reduces PKM2 expression and fibrosisPreclinical50% reduction in lesion fibrosis in animal modelsPotential hematological side effects
PFK-015PFKFB3 inhibition destabilizes β-cateninPreclinical60% inhibition of endometrial cell migrationRisk of erythrocyte dysfunction
AlisertibAURKA inhibition suppresses ERβ expression and glycolysisPreclinical40% reduction in peritoneal lactate levels in micePotential mitotic disruption
GNE 2861PAK5 inhibition blocks PKM2 phosphorylationPreclinical70% lesion size reduction in PAK5-KO miceUnconfirmed neurological effects
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

Kim, B.-S.; Kim, B.; Yoon, S.; Park, W.; Bae, S.-J.; Joo, J.; Kim, W.; Ha, K.-T. Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches. Pharmaceuticals 2025, 18, 813. https://doi.org/10.3390/ph18060813

AMA Style

Kim B-S, Kim B, Yoon S, Park W, Bae S-J, Joo J, Kim W, Ha K-T. Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches. Pharmaceuticals. 2025; 18(6):813. https://doi.org/10.3390/ph18060813

Chicago/Turabian Style

Kim, Bo-Sung, Bosung Kim, Seyeong Yoon, Wonyoung Park, Sung-Jin Bae, Jongkil Joo, Wonnam Kim, and Ki-Tae Ha. 2025. "Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches" Pharmaceuticals 18, no. 6: 813. https://doi.org/10.3390/ph18060813

APA Style

Kim, B.-S., Kim, B., Yoon, S., Park, W., Bae, S.-J., Joo, J., Kim, W., & Ha, K.-T. (2025). Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches. Pharmaceuticals, 18(6), 813. https://doi.org/10.3390/ph18060813

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