International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Over the past decade, a number of studies have shown that women with endometriosis are at increased risk for developing various diseases as comorbidities, including asthma (1), various types of cancer, such as ovarian and breast cancer, as well as cutaneous melanoma (2), cardiovascular and psychiatric diseases (3,4), hypothyroidism and fibromyalgia (1). In addition, other studies have reported an association between different autoimmune diseases, including autoimmune thyroid disorder, Crohn's disease and ulcerative colitis, Sjogren's syndrome (SS), systemic lupus erythematosus (SLE), multiple sclerosis, ankylosing spondylitis (AS), coeliac disease and rheumatoid arthritis (RA), and an increased risk for endometriosis (5-9).
Endometriosis is a common yet enigmatic inflammatory, multifactorial, estrogen-dependent gynecological condition with an unknown etiology, affecting up to 10% of women of reproductive age worldwide and 9 million women in the USA (10). Endometriosis is characterized by the presence of ectopic endometrial tissue (endometrial glands and stroma) on other organs, predominantly external to the uterine cavity and most commonly in the pelvic cavity, due to the ectopic localization of endometrial cells. Endometriosis can appear as peritoneal lesions, ovarian endometriotic cysts and deeply infiltrative endometrial lesions (11). Furthermore, endometriosis is associated with chronic pelvic pain, dysmenorrhea, dyspareunia, heavy or irregular menstrual bleeding, urinary tract symptoms, subfertility and infertility in 30% of patients (11,12). Endometriosis-associated gene polymorphisms, epigenetic modifications (such as methylation, acetylation, phosphorylation and modifications to the histone proteins associated with DNA packaging in chromatin), proinflammatory factors (including cytokines like IL-1β, IL-6, IL-8 and TNF-α, as well as prostaglandins), hormonal factors (including an imbalance between estrogen and progesterone) and environmental factors (including organic pollutants, polyhalogenated aromatic hydrocarbons, tobacco, heavy metals and pesticides) contribute to the development of the disease and trigger its specific clinical symptoms (11,12). Nowadays, a large amount of evidence indicates that dysregulation of the immune system leads to chronic inflammatory response in the ectopic endometrium, thus resulting in the subsequent development of endometriosis (13,14).
According to previous studies, women with endometriosis are at a higher risk for developing a concomitant autoimmune disease, considering the deregulation of the immune system observed in these patients (2,4,5,10,12). In this context, both proinflammatory and mitogenic proteins from endometriotic lesions, including cytokines (RANTES, IL-1, IL-6, TNF-a) and VEGF, as well as associated activated immune cells, such as pelvic macrophages and lymphocytes, can cause an imbalance in the immune system, which contributes to the enhanced inflammatory reaction that is associated with the development of endometriosis (15). Accordingly, functional abnormalities in almost all types of immune cells have been identified in endometriosis, including neutrophils, macrophages, natural killer (NK) cells, B and T lymphocytes, as well as dendritic cells (DCs) (13). Moreover, a series of anti-nuclear, anti-phospholipid and anti-endometrial antibodies are produced in endometriosis (14). This link of endometriosis with aberrant immune responses and the subsequent development of autoimmune diseases has led to the suggestion that endometriosis itself may be classified as a typical autoimmune disorder (14,16). Alternatively, another concept suggests that an underlying abnormal immune response may contribute to the development of endometriosis (5).
Recently, Chen et al (17) attempted to clarify whether patients with endometriosis have a higher risk of developing antiphospholipid syndrome (APS), by conducting a retrospective cohort study. It was demonstrated, for the first time to the best of our knowledge, that patients with endometriosis have a higher risk (2.84-fold) of developing subsequent APS compared with women without endometriosis (17). However, the existing information refers only to epidemiological and clinical aspects of this detected association. APS is a systemic autoimmune disease characterized by the occurrence of vascular or arterial thromboses at different localizations, including the brain, heart, lungs, kidneys and limbs, obstetric complications in women of childbearing age including miscarriages, recurrent fetal deaths and premature birth, and the presence of circulating antiphospholipid (aPL) antibodies (18). As an autoimmune disease, APS is developed by an interplay of genetic and antigenic factors, while specific autoantibodies are present and used for its diagnosis, such as lupus anticoagulant, anti-cardiolipin (aCL) antibodies, anti-β2 glycoprotein-I (β2GPI) antibodies and anti-phosphatidylserine/prothrombin antibodies (19,20). The genetic predisposition of APS has been demonstrated by studies in monozygotic twins, gene-association and genome-wide association studies (GWAS) as well as whole-exome sequencing (WES) approaches (21-25). Importantly, APS can occur as a single disorder where there is no clinical or laboratory evidence of another underlying disease, called ‘primary APS’ (PAPS), but it can also be associated with another autoimmune disease, and referred to in this case as ‘secondary APS’ (26). Consequently, APS may, in part, share a common mechanism for disease development or progression with other autoimmune or immunity-related diseases.
The recent data regarding the increased risk of subsequent APS in women with endometriosis posed an interesting question concerning the putative role of shared genetic factors in the co-occurrence of both conditions (17). Notably, our previous studies have focused on the delineation of the genetic components that are involved in the co-occurrence of endometriosis with various autoimmune diseases, including SLE (27), RA (7), AS (8), SS (9), as well as psoriasis and psoriatic arthritis (28). In the present review, prompted by the findings of Chen et al (17), the present review sought to delineate the genetic basis of the co-occurrence of endometriosis with APS by systematically searching the literature for genes involved in the development of both diseases, aiming to shed light on the functional significance of the shared polymorphisms and unravel the underlying molecular and pathogenetic mechanisms of these diseases.
In the current review, a potential shared genetic background regarding the co-occurrence of endometriosis and APS was investigated. To the best of our knowledge, the present review is the first study in the literature focusing on the genetic basis of the subsequent development of APS in patients with endometriosis. The present review included case-control, GWAS, whole-genome sequencing and WES studies, as well as systematic reviews and cross-sectional studies. Notably, case report series, editorial letters, expert opinions and conference abstracts were excluded. The articles included were written in the English language and published from 2000-2025. Five electronic databases were used, namely PubMed (https://pubmed.ncbi.nlm.nih.gov/), PubMed Central (https://pmc.ncbi.nlm.nih.gov/), Google Scholar (https://scholar.google.com/), Web of Science (https://www,webofscience.com/wos/) and MEDLINE (https://www.nlm.nih.gov/medline/medline_home.html/). The keywords used for the search were as follows: ‘Endometriosis’, ‘antiphospholipid syndrome’, ‘antiphospholipid antibodies’, ‘genetics’, ‘gene polymorphisms’, ‘association studies’ and ‘GWAS’. Two authors worked individually to extract the data following the specified criteria and no disagreements arose during this process.
Accordingly, certain genes were identified as potential risk factors for developing endometriosis and APS, which are involved in inflammatory and/or interferon (IFN)-inducible processes, immune system dysregulation and thrombosis (Fig. 1). The identified gene-related risk factors included the human leukocyte antigen (HLA)-DQB1*0301 (29,30) and HLA-DQA1*0301 alleles (29,31), the 4G/5G polymorphism of the serpin family E member 1 (SERPINE1) gene [encoding plasminogen activator inhibitor type 1 (PAI-1) protein] (32,33), methylenetetrahydrofolate reductase (MTHFR) rs1801133 (34,35), signal transducer and activator of transcription 4 (STAT4) rs7574865 (36,37) and toll-like receptor 4 (TLR4) rs4986790 (38,39) single nucleotide polymorphisms (SNPs). The shared gene polymorphisms and the function of the respective genes are demonstrated in Table I.
Table IOverview of genetic polymorphisms associated with both endometriosis and APS, as confirmed by gene-association and/or genome-wide association studies. |
The HLA genes encode proteins that bind antigen peptides and present them to T cells, and also play key roles in the immune response by interacting with the complement system and restricting the recognition of antigenic peptides by T cells and their involvement in cellular immunity (40). The HLA-DQB1*0301 allele of the HLA class II gene DQB1 has been associated with increased susceptibility for both endometriosis and APS (29,30). The association between endometriosis and the genes encoding the protein components of the HLA system has not yet been fully elucidated. However, considering that the suppression of cellular immunity in the peritoneal space causes endometriosis, it has been hypothesized that particular HLA gene alleles may signal these cells to decrease their cytotoxic properties (30). A notably higher frequency of the HLA-DQB1*0301 allele has been observed in patients with endometriosis, but the underlying mechanism of this association has not been fully clarified yet (30,41). The DQB1*0301 allele has also been associated with the development of APS, as well as with the presence of aCL antibodies in PAPS (29). In fact, it has been suggested that DQB1*0301 among other HLA alleles, such as HLA-DRB1*04, DRB1*07, DRB1*1302, DRw53, DQA1*0102 and DQA1*0201, determines the susceptibility to the production of aPL antibodies, which are responsible for the clinical manifestations of APS (29). Notably, HLA-DQA1*0301 allele was also found to be associated with both endometriosis and APS (29,31); this allele is associated with an increased production of β2GPI antibodies in patients with APS, thus leading to thrombotic events and pregnancy morbidity (42).
The SERPINE1 gene (coding for PAI-1) encodes a monomeric glycoprotein that is a fast-acting inhibitor of plasminogen activation, belonging to the serine protease inhibitor superfamily and regulating the fibrinolytic pathway (32). Genetic alterations of SERPINE1 have been considered as risk factors for the development of various diseases, including thrombosis (43). A common insertion/deletion SNP in the SERPINE1 gene, located 675 bp upstream from the start codon, induces the generation of two alleles containing either 4 or 5 sequential guanosines, with individuals carrying homozygosity in the deletion allele (4G/4G) having increased gene transcriptional activity and markedly elevated plasma PAI-1 protein activity compared with those in 5G/5G individuals (44,45). The 4G/5G polymorphism in the promoter region of SERPINE1 has been associated with the development of endometriosis and APS (32,33). Particularly, the 4G/4G and the 4G/5G genotypes, as well as the 4G allele of this polymorphism of the SERPINE1 gene, were found to be associated with an increased susceptibility for endometriosis. Furthermore, the 4G allele was also associated with hypofibrinolysis, which was observed at a notably higher frequency in women with endometriosis compared with that in women without endometriosis (32). Notably, it has been suggested that persistence of a fibrin matrix as a result of hypofibrinolysis could mediate the initiation of endometriotic lesions in the peritoneal cavity (32). Moreover, the presence of the 4G allele has been associated with higher plasma levels of PAI-1 and increased risk for thrombosis in patients with PAPS, compared with those in healthy controls (33).
The MTHFR gene encodes the 77-kDa MTHFR protein, which catalyzes the irreversible reduction of 5-10-MTHF to 5-methylTHF, thus representing an essential enzyme in folate and homocysteine metabolism (46) and playing an important role in DNA methylation and synthesis (46). The missense MTHFR rs1801133 (Ala222Val, C677T) SNP, located in exon 4, has been shown to be associated with both endometriosis and APS (29,30). The risk allele ‘T’ influences the enzymatic activity by producing a thermally unstable protein, which prevents optimal functioning of the enzyme at temperatures >37˚C (46) and leads to elevated plasma levels of homocysteine. In endometriosis, elevated homocysteine, along with oxidative stress, may contribute to impaired blood flow and reduced fertility (47). Notably, the increased homocysteine levels in combination with vascular inflammation and injury may lead to the development of cardiovascular disease, which is a known comorbidity of endometriosis (3,48). It is worth noting that the accumulation of high levels of cytotoxic homocysteine in the circulating blood may lead to damage of the inner wall of blood vessels. Consequently, the coagulation cascade reactions can be activated and the risk for thrombosis and ultimately APS is increased (34).
The STAT4 gene is located on chromosome 2q32.2-2q32-3(49), and encodes a transcription factor that is expressed in activated peripheral blood monocytes, macrophages, myeloid cells, DCs and T cells, and plays pivotal roles in the differentiation and proliferation of both T helper (Th) 1 and Th17 cells (49,50). Combined evidence based on gene association studies, GWAS and meta-analyses has shown an unambiguous association between STAT4 rs7574865 SNP and various autoimmune diseases, as well as chronic inflammatory diseases (51). It has been suggested that this variant of STAT4 may be involved in the regulation of the balance between the IL-12 and IL-23 effect, thus leading to inflammatory diseases through the dysregulation of the Th1 vs. Th17 differentiation (51). In addition, in silico studies have highlighted the regulatory function of this SNP in gene expression of STAT4, considering its location near both distal enhancers and an important genome regulatory factor, CCCTC-binding factor, which represents a highly conserved zinc finger protein with key roles in genome organization and gene expression, influencing both gene activation and repression (52).
In endometriosis, an increased frequency of the TT genotype of the rs7574865 (G/T) SNP has been observed in women suffering from minimal or mild endometriosis compared with that in women without endometriosis (36). Furthermore, it has been suggested that this SNP may impair either the gene expression or mRNA splicing of the STAT4 gene, thereby playing an important role in IFN signaling (36). Furthermore, the role of the rs7574865 SNP may be strengthened, considering both the strong association of the T allele with Th1 responses and the association between Th1 responses and deep infiltration in pelvic endometriosis (53). Notably, a positive association between the T allele of STAT4 rs7574865 and APS has been reported; moreover, this association was still observed after the stratification of patients with APS by the clinical manifestations of this disorder, including thrombosis or obstetric complications (37).
TLRs are pattern recognition receptors (PRRs), which form a family of transmembrane protein molecules that play a crucial role in the regulation of immune and inflammatory responses (54). TLRs activate cellular signaling pathways to induce immune-response genes, including genes encoding cytokines, chemokines and other immune mediators, and represent a host defense mechanism against infections and tissue damage, leading to the secretion of various inflammatory cytokines (55). TLR4, a type I transmembrane glycoprotein, is a PRR that mainly recognizes the lipopolysaccharide (LPS) of Gram-negative bacteria as well as structures from fungal and mycobacterial pathogens, and activates innate immunity through the production of cytokines, such as IL-1, IL-6, IL-8, TNF-α and MIP (56,57). TLR4 is composed of three domains, an extracellular, a transmembrane and a cytoplasmic domain, and was found to be involved in the pathogenesis of infectious (i.e. gram-negative infections, periodontitis and septic shock), inflammatory (obesity, liver and kidney disease, cardiovascular and neurodegenerative diseases) and autoimmune diseases (SLE, RA, MS, SSc, PS, SS and APS) (45,58).
The TLR4 rs4986790 SNP results in an aspartic acid to glycine substitution at position 299 (D299G), which alters the formation of the TLR4/myeloid differentiation factor 2 complex (59). This SNP has been associated with the development of both endometriosis and APS (25,38). In particular, the involvement of TLR4 in endometriosis has been linked to its regulatory role in the activation of immune and inflammatory responses (60). TLR4 is expressed in endometrial and endometriotic epithelial cells in women with endometriosis, and alterations in its expression levels or function have been detected in the endometrium and pelvic environment (38,60). The risk allele ‘G’ of the rs4986790 SNP was found to affect the function of TLR4. Particularly, the ‘G’ allele of the TLR4 (A896G) rs4986790 SNP was found to result in hypo-responsiveness of the receptor, thus resulting in peritoneal inflammation as suggested by Latha et al (38). Under this condition, the molecular microenvironment may become favorable for the endometrial cells to adhere to the pelvic peritoneum, thus leading to the initiation of endometriosis (38). Previous findings based on molecular dynamics simulations showed that the rs4986790 SNP may abrogate the stability of the hexamer complex that is crucial for LPS recognition and antibacterial immune response, being necessary to dimerize the cytoplasmic domain of the TLR4 protein and ultimately result in compromised TLR4 signaling (61). Notably, an eight-fold increase of endometriosis risk was seen in women who are carriers of the GG genotype (38). Regarding APS, elevated levels of TLR4 in the cell surface of peripheral blood mononuclear cells, as well as in monocytes, were observed among patients with PAPS compared with those in healthy controls (62). As suggested previously, the activation of the TLR pathway and TLR4 reactivity can lead to vascular thrombosis in patients with PAPS (45). Pierangeli et al (39) suggested that, in patients with APS, the risk allele ‘G’ of the rs4986790 SNP confers a higher susceptibility to prothrombotic endothelial activation mediated by aPL antibodies.
To the best of our knowledge, the present review is the first study attempting to delineate the genetic basis of the co-occurrence of endometriosis and APS. Although only a few shared gene polymorphisms for these disorders were identified, the present review highlighted a number of loci involved in inflammatory and/or IFN-inducible processes, immune system deregulation and thrombosis. Genetic factors are important in the development of both endometriosis and APS, as has been demonstrated previously by the familiar occurrence of these health conditions and their association with various gene polymorphisms. The identification of further genetic factors associated with these diseases is of emerging interest and may help to better delineate the mechanisms leading to their clinical association. These results are important not only for future genetic and clinical studies, but also for directing future function/structure studies on the molecular cause of endometriosis and APS. Moreover, the increasing number of disease-associated variants may contribute to the development and validation of gene risk scores for both diseases, thus improving the risk assessment and tailored management of these disorders.
Further exploration of immune cell signatures and/or cytokine profiles has emphasized the pivotal role of immune dysregulation in the immune-mediated mechanisms leading to endometriosis and APS. The specific immune cell signature of APS is characterized by activated neutrophils and an enhanced type I IFN response, while the involvement of other types of immune cells, including DCs, T and B cells, monocytes and NK cells has been also reported in the pathogenesis of APS (63,64). In endometriosis, distinct immune cells signatures have been found in the peritoneal cavity, including increased numbers of CD8+ T cells and activated NK cells, along with follicular Th cells. By contrast, M2 macrophages and resting mast cells were observed at low levels in eutopic endometria. Moreover, activated CD4+ T and B cells form part of the immune signatures associated with endometriosis (65,66), while neutrophils, macrophages, NK cells and DCs also serve a role in endometriosis (13). Cytokine profiles in APS often show an imbalance, with increased levels of pro-inflammatory cytokines, such as TNF-α and IL-6, and potentially decreased levels of anti-inflammatory cytokines, such as IL-10. Furthermore, notably elevated levels of IL-7, IL-8, IL-10, IL-16, IL-17 and IL-12/IL-23p40 and decreased levels of IL-22 and IL-31 have been detected in patients with APS compared with those in healthy controls (67,68). In women with endometriosis, elevated levels of pro-inflammatory cytokines are frequently observed in both peritoneal fluid and serum. Pro-inflammatory cytokines, such as IL-1β, IL-6, IL-8 and TNF-α, have been found to be increased in the peritoneal fluid of women with endometriosis, while some anti-inflammatory cytokines, such as IL-10 and IL-33, may also appear at elevated levels in endometriosis. IL-25 (also known as IL-17E), IL-33 and thymic stromal lymphopoietin, which are involved in Th2 cell development, are increased in the peripheral blood, endometrium and peritoneal fluid of patients with endometriosis compared with women without endometriosis. Furthermore, women with advanced endometriosis (stage 3-4) exhibit markedly increased levels of IL-1α and IL-6 in the endometrial fluid compared with those in women without endometriosis (69,70).
In recent years, genomic and epigenomic research in endometriosis and APS has grown (11,12,21,23,25,71-73), driven by novel high-throughput technologies that may identify new potential signatures and therapeutic targets for further exploration in detail. Notably, Murrin et al (74) performed a systematic analysis of the contribution of genetics to multimorbidity based on representative primary care data on 72 chronic diseases, involving individuals aged ≥65 years from two large primary-care databases. All diseases were compared between each other in pairs. The study concluded that most pairs of chronic conditions show evidence of a shared genetics background and shared pathogenetic mechanisms. Therefore, it was suggested that the identified patterns of shared genetics may provide a foundation for future multimorbidity research. Importantly, the shared genetic polymorphisms have potential implications for therapeutic interventions and a better clinical management of the patients, as further analysis of gene expression patterns and future advanced pharmacogenomic approaches may refine personalized treatment strategies for women with both conditions (74).
In a retrospective cohort study of 50,078 women with endometriosis conducted by Chen et al (17), patients showed a notably increased risk of APS regardless of the presence or absence of SLE. This is an interesting but unexpected finding, taking into account that some of the shared gene polymorphisms identified in the present study, such as the MTHFR rs1801133 and STAT4 rs7574865 SNPs also represent SLE-associated risk factors (27,75-77). Notably, STAT4 rs7574865, apart from its association with the co-occurrence of endometriosis and APS, has been also associated with the co-occurrence of endometriosis with SLE, RA, AS and SS (78).
There are a few limitations to be considered in the present study. Firstly, there is a definite epidemiological lack of replication studies thus far in the literature, concerning the development of APS in women with endometriosis in different racial and/or ethnic populations. Secondly, there is a lack of information in the literature about the role of the already identified gene polymorphisms in women with endometriosis and APS of different ethnic origins. This is an important point, taking into account the differential level of contribution of the various polymorphisms involved in the development of these disorders according to the ancestry of the populations under examination (71,79). In this framework, in a previous investigation, we showed that the availability of a number of endometriosis-associated SNPs (302 SNPs were examined) provided the opportunity for the study of this condition through SNP genomic disease genomic ‘grammar’ (DGG). Thus, the genomic pedigree of endometriosis was identified using its DGG through the reported SNPs for five major groups of the world human population, including Europeans, Africans, Americans, East Asians and South Asians, and notable ‘key’ genetic targets of endometriosis were revealed with the evidence of population-based heterogeneity (71). Thirdly, the search strategy was restricted to the English language only; therefore, there is a possibility of excluding eligible studies published in other languages. Furthermore, the source of the inter-individual susceptibility to endometriosis or APS does not lie exclusively in genetics, considering that epigenetics mechanisms, such as methylation, acetylation, phosphorylation and modifications to the histone proteins associated with DNA packaging in chromatin, or others that have been analyzed in APS, are now widely believed to participate also in the susceptibility to these diseases (70,80,81). Regarding APS, hypomethylation of ETS1 and EMP2 genes in APS neutrophils, downregulation of miR-19b and miR-20a, as well as significant reduction in methylation of the IL-8 promoter and significantly increased methylation of the tissue factor (F3) gene, have been reported so far (80,81). Thus, the role of potential shared epigenetic factors has to be investigated in future studies, considering that this issue was beyond the scope of the present preliminary study.
Despite the high number of environmental exposure factors identified for endometriosis and APS, no common factors have been reported so far. Notably, most endometriosis-associated environmental factors refer to endocrine disruptors and organic pollutants, such as polyhalogenated aromatic hydrocarbons, tobacco smoking and exposure to heavy metals, pesticides and organohalogen compounds (82,83). By contrast, the environmental triggers for APS refer to bacteria, viruses, parasites, vaccines and drugs (such as antiepileptic, antipsychotic, antihypertensive, antiarrhythmic drugs and antibiotics), as well as other factors that are potentially capable of inducing a variety of pathogenic aPL antibodies, and causing thrombosis, pregnancy loss and APS through a range of mechanisms (84).
In conclusion, the observed association between endometriosis and APS suggests that clinicians and scientists conducting research on endometriosis need to be aware of potential subsequent APS when endometriosis is diagnosed. Thus, as suggested recently, clinicians should review thoroughly the symptoms and assess risk factors for APS when evaluating women with endometriosis (17). A better functional characterization of the disease-associated genes may lead to the detection of new therapeutic targets for pharmaceutical intervention and ultimately result in improved therapeutic management for the patients by designing and producing monoclonal antibodies targeting selected genes, specific inhibitors or mimicking molecules. Notably, transcriptomic profiling and analysis have allowed for improved understanding of the mechanisms underlying endometriosis and have created opportunities for drug repurposing (85). As an example, fenoprofen, an uncommonly prescribed NSAID, was the top therapeutic candidate for further investigation, which was tested in an established rat model of endometriosis and successfully alleviated endometriosis-associated vaginal hyperalgesia (85). In the same framework, the identification of altered transcriptome signatures in samples from patients with APS and analysis using bioinformatics tools have significantly enhanced the understanding of APS pathology and led to the identification of potential therapeutic targets related to the role of the IFN pathway in this disease (81). Particularly, the upregulation of genes associated with IFN pathways suggested the potential for targeted therapeutic interventions aimed at modulating the IFN signaling pathway in patients with APS (81). Moreover, the distinct gene signatures associated with different thrombosis types and disease manifestations suggested the possibility of further tailored treatment strategies based on the patient's thrombotic profile (81). Therefore, integrating clinical and molecular data using high-throughput technologies, in combination with the employment of computational and bioinformatics tools, can advance precision medicine for patients with endometriosis and APS.
Not applicable.
Funding: No funding was received.
Not applicable.
MIZ, TBT and GNG designed the current study, searched the literature and drafted the manuscript. MIZ, BCT, GB and DAS analyzed and organized the data. TBT, BCT, GB and DAS critically revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
MIZ, TBT, BCT, GB and GNG declare that they have no competing interests. DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article.
|
Sinaii N, Cleary SD, Ballweg ML, Nieman LK and Stratton P: High rates of autoimmune and endocrine disorders, fibromyalgia, chronic fatigue syndrome and atopic diseases among women with endometriosis: a survey analysis. Hum Reprod. 17:2715–2724. 2002.PubMed/NCBI View Article : Google Scholar | |
|
Kvaskoff M, Mu F, Terry KL, Harris HR, Poole EM, Farland L and Missmer SA: Endometriosis: A high-risk population for major chronic diseases? Hum Reprod Update. 21:500–516. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Rafi U, Ahmad S, Bokhari SS, Iqbal MA, Zia A, Khan MA and Roohi N: Association of inflammatory markers/cytokines with cardiovascular risk manifestation in patients with endometriosis. Mediators Inflamm. 2021(3425560)2021.PubMed/NCBI View Article : Google Scholar | |
|
Surrey ES, Soliman AM, Johnson SJ, Davis M, Castelli-Haley J and Snabes MC: Risk of developing comorbidities among women with endometriosis: A retrospective matched cohort study. J Women's Health (Larchmt). 27:1114–1123. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Shigesi N, Kvaskoff M, Kirtley S, Feng Q, Fang H, Knight JC, Missmer SA, Rahmioglu N, Zondervan KT and Becker CM: The association between endometriosis and autoimmune diseases: A systematic review and meta-analysis. Hum Reprod Update. 25:486–503. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Yin Z, Low HY, Chen BS, Huang KS, Zhang Y, Wang YH, Ye Z and Wei JCC: Risk of ankylosing spondylitis in patients with endometriosis: A population-based retrospective cohort study. Front Immunol. 13(877942)2022.PubMed/NCBI View Article : Google Scholar | |
|
Zervou MI, Vlachakis D, Papageorgiou L, Eliopoulos E and Goulielmos GN: Increased risk of rheumatoid arthritis in patients with endometriosis: Genetic aspects. Rheumatology (Oxford). 61:4252–4262. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Zervou MI, Papageorgiou L, Vlachakis D, Spandidos DA, Eliopoulos E and Goulielmos GN: Genetic factors involved in the co-occurrence of endometriosis with ankylosing spondylitis (review). Mol Med Rep. 27(96)2023.PubMed/NCBI View Article : Google Scholar | |
|
Zervou MI, Tarlatzis BC, Grimbizis GF, Spandidos DA, Niewold TB and Goulielmos GN: Association of endometriosis with Sjögren's syndrome: Genetic insights (review). Int J Mol Med. 53(20)2024.PubMed/NCBI View Article : Google Scholar | |
|
As-Sanie S, Mackenzie SC, Morrison L, Schrepf A, Zondervan KT, Horne AW and Missmer SA: Endometriosis: A review. JAMA. 334:64–78. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Symons LK, Miller JE, Kay VR, Marks RM, Liblik K, Koti M and Tayade C: The immunopathophysiology of endometriosis. Trends Mol Med. 24:748–762. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Zondervan KT, Becker CM, Koga K, Missmer SA, Taylor RN and Viganò P: Endometriosis. Nat Rev Dis Primers. 4(9)2018.PubMed/NCBI View Article : Google Scholar | |
|
Abramiuk M, Grywalska E, Małkowska P, Sierawska O, Hrynkiewicz R and Niedźwiedzka-Rystwej P: The role of the immune system in the development of endometriosis. Cells. 11(2028)2022.PubMed/NCBI View Article : Google Scholar | |
|
Eisenberg VH, Zolti M and Soriano D: Is there an association between autoimmunity and endometriosis? Autoimmun Rev. 11:806–814. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Lebovic DI, Mueller MD and Taylor RN: Immunobiology of endometriosis. Fertil Steril. 75:1–10. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Matarese G, De Placido G, Nikas Y and Alviggi C: Pathogenesis of endometriosis: Natural immunity dysfunction or autoimmune disease? Trends Mol Med. 9:223–228. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Chen Z, Cui R, Wang SI, Zhang H, Chen M, Wang Q, Tong Q, Wei JCC and Dai SM: Increased risk of subsequent antiphospholipid syndrome in patients with endometriosis. Int J Epidemiol. 54(dyae167)2024.PubMed/NCBI View Article : Google Scholar | |
|
Clark KEN and Giles I: Antiphospholipid syndrome. Medicine. 46:118–125. 2018. | |
|
Sciascia S, Amigo MC, Roccatello D and Khamashta M: Diagnosing antiphospholipid syndrome: ‘Extra-criteria’ manifestations and technical advances. Nat Rev Rheumatol. 13:548–560. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Rodríguez-García V, Ioannou Y, Fernández-Nebro A, Isenberg DA and Giles IP: Examining the prevalence of non-criteria anti-phospholipid antibodies in patients with anti-phospholipid syndrome: A systematic review. Rheumatology (Oxford). 54:2042–2050. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Celia AI, Galli M, Mancuso S, Alessandri C, Frati G, Sciarretta S and Conti F: Antiphospholipid syndrome: Insights into molecular mechanisms and clinical manifestations. J Clin Med. 13(4191)2024.PubMed/NCBI View Article : Google Scholar | |
|
Ravindran V, Rajendran S and Elias G: Primary antiphospholipid syndrome in monozygotic twins. Lupus. 22:92–94. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Lopez-Pedrera C, Barbarroja N, Patiño-Trives AM, Collantes E, Aguirre MA and Perez-Sanchez C: New biomarkers for atherothrombosis in antiphospholipid syndrome: Genomics and epigenetics approaches. Front Immunol. 10(764)2019.PubMed/NCBI View Article : Google Scholar | |
|
Zubrzycka A, Zubrzycki M, Perdas E and Zubrzycka M: Genetic, epigenetic, and steroidogenic modulation mechanisms in endometriosis. J Clin Med. 9(1309)2020.PubMed/NCBI View Article : Google Scholar | |
|
Barinotti A, Radin M, Cecchi I, Foddai SG, Rubini E, Roccatello D, Sciascia S and Menegatti E: Genetic factors in antiphospholipid syndrome: Preliminary experience with whole exome sequencing. Int J Mol Sci. 21(9551)2020.PubMed/NCBI View Article : Google Scholar | |
|
Gómez-Puerta JA and Cervera R: Diagnosis and classification of the antiphospholipid syndrome. J Autoimmun. 48-49:20–25. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Zervou MI, Matalliotakis M and Goulielmos GN: Comment on ‘Risk of systemic lupus erythematosus in patients with endometriosis: A nationwide population-based cohort study’. Arch Gynecol Obstet. 305:543–544. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Goulielmos GN, Tarlatzi TB, Grimbizis GF, Spandidos DA, Niewold TB, Bertsias G, Tarlatzis BC and Zervou MI: Co-occurrence of endometriosis with psoriasis and psoriatic arthritis: Genetic insights (review). Exp Ther Med. 30(171)2025.PubMed/NCBI View Article : Google Scholar | |
|
Domenico Sebastiani G, Minisola G and Galeazzi M: HLA class II alleles and genetic predisposition to the antiphospholipid syndrome. Autoimmun Rev. 2:387–394. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Ishii K, Takakuwa K, Kashima K, Tamura M and Tanaka K: Associations between patients with endometriosis and HLA class II; the analysis of HLA-DQB1 and HLA-DPB1 genotypes. Hum Reprod. 18:985–989. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Zong L, Pan D, Chen W, He Y, Liu Z, Lin J and Xu A: Comparative study of HLA-DQA1 and HLA-DRB1 allele in patients with endometriosis and adenomyosis. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 19:49–51. 2002.PubMed/NCBI(In Chinese). | |
|
Bedaiwy MA, Falcone T and Casper RF: Plasminogen activator inhibitor (PAI) and thrombin activatable fibrinolysis inhibitor (TAFI) Polymorphisms in women with endometriosis. Fertil Steril. 84 (Suppl 1):S189–S190. 2005. | |
|
Aisina RB, Mukhametova LI, Ostryakova EV, Seredavkina NV, Patrushev LI, Patrusheva NL, Reshetnyak TM, Gulin DA, Gershkovich KB, Nasonov EL and Varfolomeyev SD: Polymorphism of the plasminogen activator inhibitor type 1 gene, plasminogen level and thromboses in patients with the antiphospholipid syndrome. Biochem Suppl Ser B Biomed Chem. 7:1–15. 2013. | |
|
Sayfutdinovich KZ, Shakarimovna AD, Rudolfovna KT and Erkinovna ZN: Association of MTHFR and MTRR genes with the development of antiphospholipid syndrome in pregnant women of Uzbek population. Eur Sci Rev. 7-8:85–87. 2016. | |
|
Delli Carpini G, Giannella L, Di Giuseppe J, Montik N, Montanari M, Fichera M, Crescenzi D, Marzocchini C, Meccariello ML, Di Biase D, et al: Homozygous C677T methylenetetrahydrofolate reductase (MTHFR) polymorphism as a risk factor for endometriosis: A retrospective case-control study. Int J Mol Sci. 24(15404)2023.PubMed/NCBI View Article : Google Scholar | |
|
Bianco B, Fernandes RFM, Trevisan CM, Christofolini DM, Sanz-Lomana CM, de Bernabe JV and Barbosa CP: Influence of STAT4 gene polymorphisms in the pathogenesis of endometriosis. Ann Hum Genet. 83:249–255. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Horita T, Atsumi T, Yoshida N, Nakagawa H, Kataoka H, Yasuda S and Koike T: STAT4 single nucleotide polymorphism, rs7574865 G/T, as a risk for antiphospholipid syndrome. Ann Rheum Dis. 68:1366–1367. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Latha M, Vaidya S, Movva S, Chava S, Govindan S, Govatati S, Banoori M, Hasan Q and Kodati VL: Molecular pathogenesis of endometriosis; Toll-like receptor-4 A896G (D299G) polymorphism: A novel explanation. Genet Test Mol Biomarkers. 15:181–184. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Pierangeli SS, Vega-Ostertag ME, Raschi E, Liu X, Romay-Penabad Z, De Micheli V, Galli M, Moia M, Tincani A, Borghi MO, et al: Toll-like receptor and antiphospholipid mediated thrombosis: In vivo studies. Ann Rheum Dis. 66:1327–1333. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Carey BS, Poulton KV and Poles A: Factors affecting HLA expression: A review. Int J Immunogenet. 46:307–320. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Ali AM, Nader MI, Al-Ghurabi BH and Mohammed AK: Association of HLA class II alleles (DRB1 and DQB1) in Iraqi women with endometriosis. Iraqi J Biotechnol. 15:42–50. 2016. | |
|
Ioannidis JP, Tektonidou MG, Vlachoyiannopoulos PG, Stavropoulos-Giokas C, Spyropoulou-Vlachou M, Reveille JD, Arnett FC and Moutsopoulos HM: HLA associations of anti-beta2 glycoprotein I response in a Greek cohort with antiphospholipid syndrome and meta-analysis of four ethnic groups. Hum Immunol. 60:1274–1280. 1999.PubMed/NCBI View Article : Google Scholar | |
|
Mansilha A, Araújo F, Severo M, Sampaio SM, Toledo T, Henriques I and Albuquerque R: The association between the 4G/5G polymorphism in the promoter of the plasminogen activator inhibitor-1 gene and deep venous thrombosis in young people. Phlebology. 20:48–52. 2005. | |
|
Dossenbach-Glaninger A, van Trotsenburg M, Dossenbach M, Oberkanins C, Moritz A, Krugluger W, Huber J and Hopmeier P: Plasminogen activator inhibitor 1 4G/5G polymorphism and coagulation factor XIII Val34Leu polymorphism: Impaired fibrinolysis and early pregnancy loss. Clin Chem. 49:1081–1086. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Islam MA, Khandker SS, Alam F, Kamal MA and Gan SH: Genetic risk factors in thrombotic primary antiphospholipid syndrome: A systematic review with bioinformatic analyses. Autoimmun Rev. 17:226–243. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Raghubeer S and Matsha TE: Methylenetetrahydrofolate (MTHFR), the one-carbon cycle, and cardiovascular risks. Nutrients. 13(4562)2021.PubMed/NCBI View Article : Google Scholar | |
|
Guedes T, Santos AA, Vieira-Neto FH, Bianco B, Barbosa CP and Christofolini DM: Folate metabolism abnormalities in infertile patients with endometriosis. Biomark Med. 16:549–557. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Luo Z, Lu Z, Muhammad C, Chen Y, Chen Q, Zhang J and Song Y: Associations of the MTHFR rs1801133 polymorphism with coronary artery disease and lipid levels: A systematic review and updated meta-analysis. Lipids Health Dis. 17(191)2018.PubMed/NCBI View Article : Google Scholar | |
|
Wu S, Wang M, Wang Y, Zhang M and He JQ: Polymorphisms of the STAT4 gene in the pathogenesis of tuberculosis. Biosci Rep. 38(BSR20180498)2018.PubMed/NCBI View Article : Google Scholar | |
|
Korman BD, Kastner DL, Gregersen PK and Remmers EF: STAT4: Genetics, mechanisms, and implications for autoimmunity. Curr Allergy Asthma Rep. 8:398–403. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Liang YL, Wu H, Shen X, Li PQ, Yang XQ, Liang L, Tian WH, Zhang LF and Xie XD: Association of STAT4 rs7574865 polymorphism with autoimmune diseases: A meta-analysis. Mol Biol Rep. 39:8873–8882. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Bravo-Villagra KM, Muñoz-Valle JF, Baños-Hernández CJ, Cerpa-Cruz S, Navarro-Zarza JE, Parra-Rojas I, Aguilar-Velázquez JA, García-Arellano S and López-Quintero A: STAT4 gene variant rs7574865 is associated with rheumatoid arthritis activity and anti-CCP levels in the western but not in the southern population of Mexico Mexico. Genes (Basel). 15(241)2024.PubMed/NCBI View Article : Google Scholar | |
|
Podgaec S, Dias Junior JA, Chapron C, de Oliveira RM, Baracat EC and Abrão MS: Th1 and Th2 ummune responses related to pelvic endometriosis. Assoc Med Bras (1992). 56:92–98. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Medzhitov R and Janeway C Jr: Innate immunity. N Engl J Med. 343:338–344. 2000.PubMed/NCBI View Article : Google Scholar | |
|
Ozato K, Tsujimura H and Tamura T: Toll-like receptor signaling and regulation of cytokine gene expression in the immune system. BioTechniques. 33 (Suppl):S66–S75. 2002.PubMed/NCBI | |
|
Poltorak A, Smirnova I, He X, Liu MY, Van Huffel C, McNally O, Birdwell D, Alejos E, Silva M, Du X, et al: Genetic and physical mapping of the Lps locus: Identification of the toll-4 receptor as a candidate gene in the critical region. Blood Cells Mol Dis. 24:340–355. 1998.PubMed/NCBI View Article : Google Scholar | |
|
Akira S, Uematsu S and Takeuchi O: Pathogen recognition and innate immunity. Cell. 124:783–801. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Kim HJ, Kim H, Lee JH and Hwangbo C: Toll-like receptor 4 (TLR4): New insight immune and aging. Immun Ageing. 20(67)2023.PubMed/NCBI View Article : Google Scholar | |
|
Yamakawa N, Ohto U, Akashi-Takamura S, Takahashi K, Saitoh S, Tanimura N, Suganami T, Ogawa Y, Shibata T, Shimizu T and Miyake K: Human TLR4 polymorphism D299G/T399I alters TLR4/MD-2 conformation and response to a weak ligand monophosphoryl lipid A. Int Immunol. 25:45–52. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Khan KN, Kitajima M, Fujishita A, Nakashima M and Masuzaki H: Toll-like receptor system and endometriosis. J Obstet Gynaecol Res. 39:1281–1292. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Anwar MA and Choi S: Structure-activity relationship in TLR 4 mutations: Atomistic molecular dynamics simulations and residue interaction network analysis. Sci Rep. 7(43807)2017.PubMed/NCBI View Article : Google Scholar | |
|
Benhamou Y, Bellien J, Armengol G, Brakenhielm E, Adriouch S, Iacob M, Remy-Jouet I, Le Cam-Duchez V, Monteil C, Renet S, et al: Role of Toll-like receptors 2 and 4 in mediating endothelial dysfunction and arterial remodeling in primary arterial antiphospholipid syndrome. Arthritis Rheum. 66:3210–3220. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Palli E, Kravvariti E and Tektonidou MG: Type I interferon signature in primary antiphospholipid syndrome: Clinical and laboratory associations. Front Immunol. 10(487)2019.PubMed/NCBI View Article : Google Scholar | |
|
Tang KT, Chen HH, Chen TT, Bracci NR and Lin CC: Dendritic cells and antiphospholipid syndrome: An updated systematic review. Life (Basel). 11(801)2021.PubMed/NCBI View Article : Google Scholar | |
|
Wu XG, Chen JJ, Zhou HL, Wu Y, Lin F, Shi J, Wu HZ, Xiao HQ and Wang W: Identification and validation of the signatures of infiltrating immune cells in the eutopic endometrium endometria of women with endometriosis. Front Immunol. 12(671201)2021.PubMed/NCBI View Article : Google Scholar | |
|
Ahn SH, Khalaj K, Young SL, Lessey BA, Koti M and Tayade C: Immune-inflammation gene signatures in endometriosis patients. Fertil Steril. 106:1420–1431.e7. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Hubben AK, Bazeley P, Swaidani S, Alarabi A, Kulkarni PP, Shim YJ, Palihati M, Barnard J and Mccrae KR: Cytokine profiling in antiphospholipid syndrome demonstrates persistent immune dysregulation and a procoagulant phenotype. Blood. 142 (Suppl 1)(S5400)2023. | |
|
Yan H, Li B, Su R, Gao C, Li X and Wang C: Preliminary study on the imbalance between Th17 and regulatory T cells in antiphospholipid syndrome. Front Immunol. 13(873644)2022.PubMed/NCBI View Article : Google Scholar | |
|
Llarena NC, Richards EG, Priyadarshini A, Fletcher D, Bonfield T and Flyckt RL: Characterizing the endometrial fluid cytokine profile in women with endometriosis. J Assist Reprod Genet. 37:2999–3006. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Oală IE, Mitranovici MI, Chiorean DM, Irimia T, Crișan AI, Melinte IM, Cotruș T, Tudorache V, Moraru L, Moraru R, et al: Endometriosis and the Role of Pro-Inflammatory and Anti-Inflammatory Cytokines in Pathophysiology: A narrative review of the literature. Diagnostics (Basel). 14(312)2024.PubMed/NCBI View Article : Google Scholar | |
|
Papageorgiou L, Andreou A, Zervou M, Vlachakis D, Goulielmos GN and Eliopoulos E: A global population genomic analysis shows novel insights into the genetic characteristics of endometriosis. World Acad Sci J. 5(12)2023. | |
|
Goulielmos GN, Matalliotakis M, Matalliotaki C, Eliopoulos E, Matalliotakis I and Zervou MI: Endometriosis research in the-omics era. Gene. 741(144545)2020.PubMed/NCBI View Article : Google Scholar | |
|
Zouein J, Naim N, Spencer DM and Ortel TL: Genetic and genomic associations in antiphospholipid syndrome: A systematic review. Autoimmun Rev. 24(103712)2025.PubMed/NCBI View Article : Google Scholar | |
|
Murrin O, Mounier N, Voller B, Tata L, Gallego-Moll C, Roso-Llorach A, Carrasco-Ribelles LA, Fox C, Allan LM, Woodward RM, et al: A systematic analysis of the contribution of genetics to multimorbidity and comparisons with primary care data. EBioMedicine. 113(105584)2025.PubMed/NCBI View Article : Google Scholar | |
|
Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, de Bakker PIW, Le JM, Lee HS, Batliwalla F, et al: STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 357:977–986. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Zhou HY and Yuan M: MTHFR polymorphisms (rs1801133) and systemic lupus erythematosus risk: A meta-analysis. Medicine (Baltimore). 99(e22614)2020.PubMed/NCBI View Article : Google Scholar | |
|
Chen L, Huang Z, Liao Y, Yang B and Zhang J: Association between tumor necrosis factor polymorphisms and rheumatoid arthritis as well as systemic lupus erythematosus: A meta-analysis. Braz J Med Biol Res. 52(e7927)2019.PubMed/NCBI View Article : Google Scholar | |
|
Zervou MI and Goulielmos GN: Comment on: Enhancing current guidance for psoriatic arthritis and its comorbidities: Recommendations from an expert consensus panel. Rheumatology (Oxford). 64:904–905. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Uthman I and Khamashta M: Ethnic and geographical variation in antiphospholipid (Hughes) syndrome. Ann Rheum Dis. 64:1671–1676. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Weeding E, Coit P, Yalavarthi S, Kaplan MJ, Knight JS and Sawalha AH: Genome-wide DNA methylation analysis in primary antiphospholipid syndrome neutrophils. Clin Immunol. 196:110–116. 2018.PubMed/NCBI View Article : Google Scholar | |
|
López-Pedrera C, Cerdó T, Jury EC, Muñoz-Barrera L, Escudero-Contreras A, Aguirre MA and Pérez-Sánchez C: New advances in genomics and epigenetics in antiphospholipid syndrome. Rheumatology (Oxford). 63 (SI):SI14–SI23. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Caporossi L, Capanna S, Viganò P, Alteri A and Papaleo B: From environmental to possible occupational exposure to risk factors: What role do they play in the etiology of endometriosis? Int J Environ Res Public Health. 18(532)2021.PubMed/NCBI View Article : Google Scholar | |
|
Matta K, Lefebvre T, Vigneau E, Cariou V, Marchand P, Guitton Y, Royer AL, Ploteau S, Le Bizec B, Antignac JP and Cano-Sancho G: Associations between persistent organic pollutants and endometriosis: A multiblock approach integrating metabolic and cytokine profiling. Environ Int. 158(106926)2022.PubMed/NCBI View Article : Google Scholar | |
|
Martirosyan A, Aminov R and Manukyan G: Environmental triggers of autoreactive responses: Induction of antiphospholipid antibody formation. Front Immunol. 10(1609)2019.PubMed/NCBI View Article : Google Scholar | |
|
Oskotsky TT, Bhoja A, Bunis D, Le BL, Tang AS, Kosti I, Li C, Houshdaran S, Sen S, Vallvé-Juanico J, et al: Identifying therapeutic candidates for endometriosis through a transcriptomics-based drug repositioning approach. iScience. 27(109388)2024.PubMed/NCBI View Article : Google Scholar |