Back to Journals » Journal of Multidisciplinary Healthcare » Volume 18

Causal Effects of Immune Cells on Reproductive Ill-Health, Including Abnormal Spermatozoa, Polycystic Ovary Syndrome and Spontaneous Abortion: Mendelian Randomization Analyses

Authors Chen S , Sun S, Zhou Z, Zhou Z , Zhang R, Song W, Xin H, Yang Q, Dai S, Huang K, Niu W, Shi H, Guo Y

Received 28 February 2025

Accepted for publication 28 May 2025

Published 6 June 2025 Volume 2025:18 Pages 3219—3232

DOI https://doi.org/10.2147/JMDH.S524949

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr David C. Mohr



Shuang Chen,1,* Shihao Sun,2,* Zihan Zhou,1,* Zhaokai Zhou,3 Ran Zhang,1 Wenyan Song,1 Hang Xin,1 Qingling Yang,1 Shanjun Dai,1 Kai Huang,1 Wenbin Niu,1 Hao Shi,1 Yihong Guo1

1Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China; 2Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China; 3Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Yihong Guo, Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People’s Republic of China, Email [email protected]

Background: Accumulative prior studies have demonstrated that immune inflammation profoundly influences reproductive disorders of mesodermal origin. However, little is known about the causal relationship between immune factors and diseases of the reproductive system.
Methods: Thorough two-sample Mendelian randomization (MR) analyses were conducted to determine the causal effects of 731 immune traits on reproductive ill-health, including abnormal spermatozoa (AS), polycystic ovary syndrome (PCOS), and spontaneous abortion (SA). Causal links were decrypted using genome-wide association study (GWAS) data. Sensitivity analyses were performed to assess the strength, heterogeneity, and horizontal pleiotropy of the results.
Results: For AS, 34 causal relationships were identified, with BAFF-R, CD20, and CD27 in the B-cell panel having protective effects against AS. A crucial causative connection between CD11c+ CD62L− monocyte%monocyte (cDC panel) and AS pathogenesis was also revealed. For PCOS, 40 causal effects were established, with CD20, CD24, and CD27 in the B-cell panel playing different roles in PCOS. CD4 on CM CD4+ (maturation stages of the T-cell panel) significantly increased the risk of PCOS. For SA, 33 causative associations were determined, and a protective effect of CCR2 (C-C chemokine receptor type 2) on CD14+ CD16+ monocytes (monocyte panel) in SA was particularly noted. The diverse functions of the CD28, CD39, and CD25 molecules in the Treg cell panel in SA were also observed.
Conclusion: This study comprehensively evaluated the causal impact of immune traits on reproductive illnesses, stressing the complex and important role of immunogenic factors in pathogenesis and highlighting a novel direction for clinical work.

Keywords: Mendelian randomization, immunity, causal inference, abnormal spermatozoa, polycystic ovary syndrome, spontaneous abortion

Introduction

The prevalence of infertility is 10–15%, with male factors accounting for approximately 40%. Reproductive disorders have emerged as an issue of concern throughout the world.1,2 The current downward trend in global fertility, with projections of a decline in the world population after a possible peak of 9.7 billion in 2064 to a world population of 8.8 billion by 2100, might have dramatic negative implications for social advancement.3 The mesodermal origin of the reproductive system, including the testes, endometrium, and ovaries, is crucial for the formation and development of the embryo, and corresponding reproductive disorders, such as abnormal spermatozoa (AS), polycystic ovary syndrome (PCOS) and spontaneous abortion (SA), cause distress and confusion in countless families.4,5 High-quality sperm is essential for the penetration and activation of oocytes. However, approximately 2% of infertile males have abnormal sperm parameters.6 Given that sperm morphology is linked to sperm motility, the ability of sperm to penetrate the cervical mucus, and the capacity of sperm to enter the oocyte zona pellucida, increases in the rates of sperm deformity may postpone natural conception and increase the likelihood of spontaneous miscarriage.7 Therefore, it is critical to investigate the pathogenesis of AS to improve clinical practice. Infertility in women is predominantly caused by PCOS, which afflicts 6–12% of women of reproductive age worldwide.8,9 In addition to having typical clinical manifestations encompassing hyperandrogenaemia, ovulatory dysfunction, and polycystic ovarian morphology, PCOS patients suffer from a wide range of endocrine metabolic dysfunctions, such as insulin resistance, as well as the accompanying potential risk for type 2 diabetes mellitus and cardiovascular disease, all of which are detrimental to physical and mental health.10–12 Despite recent advances suggesting that PCOS is a complex disorder influenced by genetic and environmental factors, its aetiology and underlying biological processes remain unclear. SA, generally referred to as miscarriage, is defined as the loss of a nonviable intrauterine pregnancy before 20 weeks of gestation and affects approximately 10–20% of pregnancies, with the majority of SA (~80%) occurring before 13 weeks of gestation.4,13–15 Successful embryo implantation and gestational processes rely on a high-quality endometrium to afford the embryo the opportunity to attach, invade, and develop, highlighting that both the endometrium and embryo quality play pivotal roles in the outcome of embryo implantation.16 The decidualized endometrium functions as a biosensor of embryo quality and, if disrupted, can lead to the implantation of embryos destined for miscarriage.17 A decreased number of p16-positive senescent cells in the endometrium has been recognized as a biomarker of miscarriage.18 Further research aiming to achieve a more thorough understanding of the causative mechanisms of miscarriages is needed, as this topic is highly important for preventing miscarriages.

In recent years, with the introduction of the “reproductive immune microenvironment (RIM)”, research on the underlying connection between the immune and reproductive systems has been in the spotlight.19 Increasing evidence suggests that the immune microenvironment is strongly associated with reproductive disorders.20–22 As the core sites for the production and growth of germ cells and fertilized oocytes, the testes, ovaries and uterus together constitute a specialized “reproductive microenvironment with immune privilege”.23–26 Nevertheless, multiple potential threats may have negligible effects on the RIM, resulting in immune infertility afflicting couples, for which a great deal of research is underway.19 Testicular macrophages promote spermatogenesis via colony-stimulating factor 127 and steer embryonic testicular development by secreting growth factors and cytokines.28 Thus, testicular macrophages are recognized as potential targets for male infertility treatment.20 The immune response of the ovary is an exciting and emerging research area that is essential for understanding the complexity of ovary-related diseases.22 As early as 1978, the hypothesis that immune cells play a pivotal regulatory role in ovarian cycle regulation and follicle number selection was proposed.29 Immune cells are pivotal in follicle development and oestrogen synthesis, ranging from phagocytosis and antigen presentation to the secretion of proteases, cytokines, chemokines, and growth factors, among others.22,30,31 In addition, studies have revealed the importance of immune cells in endometrial tolerance and early placental development.32–34 Altered numbers or dysfunctions of uterine immune cell populations, especially natural killer cells and regulatory T cells, have also been demonstrated to be strongly involved in miscarriage.32 Decidual NK cells, which are involved in vascular remodelling and contribute to trophoblast invasion, are also implicated in miscarriages and other pregnancy failures when suppressed.35 To date, investigations into the effects of immunological factors on the pathogenesis of reproductive health problems have not been widespread or sufficiently thorough. More refined and holistic studies are urgently needed to explore the potential causal relationships between immunologic factors and reproductive disorders. Exploration of the risk or protective factors involved in these diseases from an immune perspective can yield information for infertility diagnosis, and potential therapeutic strategies such as treatment to suppress immune cells may be of clinical utility.

Mendelian randomization (MR) is a unique tactic of genetic epidemiology research designed to assess the causal effects of exposures on clinical outcomes in specific diseases.36,37 This strategy employs genetic variants strongly linked to certain exposures as instrumental variables (IVs), offering reliable and impartial estimations of genotypic determinations during conception.38 To perform MR analyses, three essential assumptions must be satisfied: first, the genetic variance should be closely related to the exposure; second, the genetic variance should not be susceptible to other confounding variables; and third, genetic IVs can influence the outcome only via the exposure.39,40 With this credible approach, investigators can more precisely and holistically probe the underlying causal influences of multiple factors on the disease, laying the groundwork for future clinical therapeutic strategies.41 Over the past decade, MR has been broadly used to deduce causality with publicly available genome-wide association study (GWAS) data.42–44

Herein, we performed a comprehensive two-sample Mendelian randomization analysis to evaluate the cause-and-effect relationships between immune traits and reproductive ill-health, including AS, PCOS, and SA. This study is the first to reveal a new underlying causality between the immune system and reproductive ill health, which has not been identified in previous studies, paving the way for the discovery of immunogenic factors in the etiopathogenesis of reproductive ill health.

Materials and Methods

Study Design

A comprehensive two-sample MR study was conducted to investigate the causal links between 731 immune traits and reproductive ill-health. Figure 1A depicts the study design alongside three indispensable MR assumptions: (1) genetic instruments are linked to the exposures, (2) genetic variants are unrelated to any confounding factors, and (3) genetic instruments influence outcomes only via risk variables.45 A graphical representation of the study design is presented in Figure 1B.

Figure 1 Overview of the study design. (A) Schematic representation of the three core assumptions underlying MR analysis: Assumption 1: The correlation hypothesis: genetic variation is directly related to exposure; Assumption 2: The Independence hypothesis: there is no connection between the genetic variant and the possible confounders between exposure and outcome; Assumption 3: The exclusion hypothesis: the genetic variation will not affect the outcome by means other than exposure. (B) Flowchart outlining the overall study design.

Abbreviations: MR, Mendelian randomization; IVs, instrumental variables; IVW, inverse-variance weighted; MR-PRESSO, MR pleiotropy residual sum and outlier.

GWAS Data Sources for Reproductive Ill-Health Status

Summary statistics for AS were obtained from the FinnGen Consortium R7 release data (https://r7.finngen.fi/). With a total of 1913 cases and 293,878 controls, GWAS data with the “abnormal spermatozoa” phenotype were retrieved. Statistics for PCOS and SA were obtained from the FinnGen Consortium R9 release data (https://r9.finngen.fi/). For PCOS, the specific phenotype code was categorized as “R9_E4_PCOS”, and a total of 1424 cases and 200,581 controls were derived. For SA, the corresponding phenotype code used in this study was “O15_ABORT_SPONTAN”, with 16,906 cases and 149,622 controls.

Immunity-Wide GWAS Data Sources

To evaluate the causal relationships between immune metrics and reproductive ill-health, we employed the most extensive GWAS data from peripheral blood immunophenotyping to date, which utilized raw genetic immune profiles from 3757 Europeans. In total, 731 immunophenotypes were identified, covering absolute cell (AC) counts (n = 118), median fluorescence intensities (MFI, n = 389, indicating surface antigen levels), morphological parameters (MP, n = 32, including forward and side scatter, reflecting cell volume, intracellular complexity, and cell-surface texture), and relative cell counts (RC, n = 192). The GWAS summary statistics of 731 immune traits are available in the GWAS Catalog (accession numbers from GCST0001391 to GCST0002121).46 Specifically, the MFI, AC, and RC metrics encompassed B cells, cDCs, mature stages of T cells, monocytes, myeloid cells, TBNK (T cells, B cells, and natural killer cells), and Treg panels, whereas the MP feature incorporated the cDC and TBNK panels.

Selection of Instrumental Variables (IVs)

A rigorous screening procedure was applied to the IVs to ensure the precision and credibility of the analyses. Drawing from published studies,46 we set a significance level threshold of 1 × 10–5 for IVs pertaining to each immune trait. This predetermined criterion continued to dictate the significance of the differences in the results for reproductive system problems. Pruning of single nucleotide polymorphisms (SNPs) was conducted via the clumping procedure of PLINK software (version v1.90), and we established a linkage disequilibrium (LD) r2 threshold below 0.1 within a 500-kb distance. As a reference panel, the 1000 Genomes Project was utilized to calculate LD r² values.47 To mitigate the risk of biased discoveries due to improperly chosen IVs, specific palindromic SNPs were excluded. To avert latent instrumental bias arising from weak instruments, the F-statistic (β2_exposure/SE2_exposure) was computed to appraise the potency of IVs, and F-statistics exceeding 10 indicated that IVs were sufficient for reliable MR analysis.48,49 These criteria not only adhered to the recommendations of previous studies but also ensured the dependability and accuracy of the IVs employed in this study.

Assessment of Causal Effects and Sensitivity Analysis

To investigate the causal effects of immune traits on reproductive disorders, we utilized several reliable MR methods built on distinctive assumptions. Primary analysis relied on the inverse-variance weighted (IVW) method,50 whereas the weighted median,51 MR‒Egger,52 and MR pleiotropy residual sum and outlier (MR-PRESSO)53 were employed to validate the robustness of the IVW results.

Thorough and methodical sensitivity analyses were also conducted to eliminate underlying violations of the MR assumptions. Heterogeneity was estimated using Cochran’s Q statistic, where a p value less than 0.05 suggested significant heterogeneity.54 In cases where the null hypothesis was rejected, implying possible heterogeneity within IVs, we applied the random-effect IVW method as an alternative to the fixed-effect IVW approach.50 Horizontal pleiotropy and potential bias due to invalid IVs were examined using MR-Egger intercept analysis.55 MR-PRESSO outlier tests were employed to investigate the possibility of horizontal pleiotropy introducing bias into the MR results. We detected genetic variations in the outliers using MR-PRESSO and reevaluated the impact estimates after their removal.53 The ‘leave-one-out’ (LOO) method validated the dependability and reliability of the results by the iteratively removing each SNP and subsequently performing MR analyses. LOO visually confirmed whether a single SNP could drive the predominant causation.52 Scatter plots revealed that the outcomes were unaffected by outliers. Funnel plots illustrated the robustness of causal inferences, with no evident heterogeneity. Additionally, we adopted MR Steiger directionality tests to determine the accuracy of the deduced causal direction.56

A strong causal relationship between immune traits and reproductive disorders could be established in this study only when the following criteria were met: 1) The IVW method indicated a significant association with a p-value <0.05; 2) The estimates derived from the IVW method were consistent with those from the other three MR methods; 3) Cochran’s Q test, MR-Egger intercept analysis, and MR-PRESSO global test showed no significant results (p > 0.05); 4) The MR-Steiger directionality test confirmed a correct causal direction; 5) The MR estimates were not significantly influenced by a single SNP in the LOO analysis, ensuring the robustness of the findings. Only immune traits that met all these criteria could be considered as potential protective or risk factors for reproductive disorders.

Statistical Analysis

All MR analyses were performed using R software (version 4.2.0). Packages, including MendelianRandomization and MR-PRESSO, were used. A comprehensive array of methods and tests has established a logical framework for estimating the causal effects between immune traits and reproductive ill health. The criteria for establishing a causal relationship have been detailed in the preceding sections. Statistical significance was defined as a two-tailed p-value <0.05, in all sensitivity analysis tests.

Results

Exploration of the Causal Effects of Immunophenotypes on AS

To assess the causal effects of immunophenotypes on AS, a two-sample MR analysis was applied. Among the 731 immunophenotypes, 34 immune traits were found to be significantly causally associated with AS, 13 of which were distributed in the B-cell panel (Figure 2). Except for IgD+ CD38dim AC (B-cell panel, odds ratio [OR]: 1.036; 95% confidence interval [CI]: 1.001–1.072; PIVW =0.042), which increased the risk of AS onset, the remaining 12 immune signatures of the B-cell panel had protective effects against AS. BAFF-R, CD20, and CD27 were highlighted in the B-cell panel, and the associated immunophenotypes were causally linked to a reduced risk of AS development, including BAFF−R on CD24+ CD27+ cells (B-cell panel, OR: 0.968; 95% CI: 0.937–0.999; PIVW =0.044), CD20 on IgD+ CD38− (B-cell panel, OR: 0.921; 95% CI: 0.860–0.986; PIVW =0.018), and CD27 on IgD− CD38− (B-cell panel, OR: 0. 929; 95% CI: 0. 870–0. 992; PIVW =0.029). Notably, the causal relationship between CD11c+ CD62L- monocyte%monocyte and AS was the most significant (cDC panel, PIVW =0.0005). With 17 SNPs as proxy predictors, an elevated CD11c+ CD62L- monocyte%monocyte was strongly correlated with a substantial increase in AS risk (OR: 1.228; 95% CI: 1.094–1.378). Similarly, other immune traits in the cDC panel, including CD11c+ monocyte%monocyte (cDC panel, OR: 1.111; 95% CI: 1.016–1.214; PIVW =0.021) and CD62L− plasmacytoid DC%DC (cDC panel, OR: 1.101; 95% CI: 1.021–1.214; PIVW =0.188), were also found to contribute to the increased risk of AS development.

Figure 2 Causal associations between immune traits and AS: Results of MR analyses using the IVW method.

Abbreviations: AS, abnormal spermatozoa; MR, Mendelian randomization; IVW, inverse-variance weighted; OR, odds ratio.

Although the IVW technique is fairly effective at determining whether an exposure causes a complicated disease, it may be influenced by weak instrumental biases. To mitigate these biases, we employed multiple MR analytical strategies to determine the reliability of the 34 causal relationships deduced by the IVW method. Causality estimates consistent with the IVW approach were also derived from the three other MR methods, namely, MR‒Egger, weighted median, and MR-PRESSO (Table S1). Interestingly, CD11c+ CD62L- monocyte%monocyte, the most salient immunophenotype identified by the IVW approach, also passed some additional sensitivity tests, showing a strong cause-and-effect relationship with AS risk (ORweighted-median: 1.24, 95% CIweighted-median: 1.04–1.47, Pweighted-median = 0.016; ORMR-PRESSO: 1.23, 95% CIMR-PRESSO: 1.07–1.40, PMR-PRESSO = 0.0025), indicating that more in-depth studies are needed (Figure S1A and Table S1). The LOO method demonstrated the absence of major SNPs in IVs that might significantly impact the results after culling, thus increasing the confidence in causality (Figure S1B). The funnel plots showed no signs of horizontal pleiotropy or heterogeneity, further supporting the dependability of the causal effect of CD11c+ CD62L- monocyte%monocyte on AS (Figure S1C).

Subsequently, a range of sensitivity analyses was conducted to test the plausibility of the results. For the 34 immune features mined using IVW, Cochran’s Q test revealed no heterogeneity among the different IVs (Table S2). MR‒Egger’s intercept test and MR-PRESSO global tests showed no potential effects of level-multiplicity. MR Steiger directionality tests affirmed the precision of the causal effects’ directionality between immune features and AS (Table S3).

Investigation of Causal Relationships Between Immunophenotypes and PCOS

Among the 731 immunophenotypes, 40 immune signatures were causally associated with PCOS (Figure 3). The maturation stages of the T cell panel were highlighted for causality in relation to the increased risk of PCOS onset, with CD4 on CM CD4+ presenting the most significant causal link to PCOS (maturation stages of T cell panel, OR: 1.171; 95% CI: 1.059–1.294; PIVW = 0.002). Similarly, HVEM on EM CD8br (maturation stages of T cell panel, OR: 1.096; 95% CI: 1.018–1.180; PIVW = 0.015) and of CCR7 expression on naive CD8b (maturation stages of T cell panel, OR: 1.097; 95% CI: 1.005–1.197; PIVW = 0.038) were identified as potential risk factors for PCOS pathogenesis. Notably, complex correlations between B cells and PCOS (11 protective associations and 6 risk associations in causality) were detected, and the CD20, CD24, and CD27 molecules in the B cell panel were stressed. In particular, the protective role of CD27 in PCOS was brought to attention, comprising CD27 on CD20− (B-cell panel, OR: 0.925; 95% CI: 0.861–0.994; PIVW = 0.035), CD27 on CD20− CD38− (B-cell panel, OR: 0.890; 95% CI: 0.803–0.986; PIVW = 0.027), among other instances. Furthermore, immune features of the cDC panel, such as DC AC (cDC panel, OR: 1.114; 95% CI: 1.010–1.228; PIVW = 0.031), CD86 on myeloid DC (cDC panel, OR: 1.134; 95% CI: 1.019–1.261; PIVW = 0. 021), and CD62L− DC%DC (cDC panel, OR: 1.073; 95% CI: 1.005–1.145; PIVW = 0.034) increased the risk of PCOS. Monocytes containing CX3CR1 on CD14+ CD16+ monocyte (monocyte panel, OR: 0.937; 95% CI: 0.880–0.997; PIVW = 0.038) and PDL−1 on CD14− CD16+ monocyte (monocyte panel, OR: 0.917; 95% CI: 0.862–0.976; PIVW = 0.007) were detected as protective immunogenic factors in PCOS patients.

Figure 3 Causal associations between immune traits and PCOS: Results of MR analyses using the IVW method.

Abbreviations: PCOS, polycystic ovary syndrome; MR, Mendelian randomization; IVW, inverse-variance weighted; OR, odds ratio.

Congruent causality estimates for the 40 causal effects were also supported by the MR‒Egger, weighted median, and MR-PRESSO analyses (Table S4). The prominent risk-predictive role of CD4 on CM CD4+ for PCOS was evidenced by additional tests (maturation stages of T cell panel, ORMR-PRESSO: 1.17, 95% CIMR-PRESSO: 1.08–1.27, PMR-PRESSO = 0.00013) (Figure S2A and Table S4), and there were no possible outliers in the IVs according to the LOO approach (Figure S2B). Funnel plots further indicated the reliability of this causality and no heterogeneity (Figure S2C). In addition, no signs of heterogeneity were found in our study using the Cochran’s Q test (Table S5). MR‒Egger’s intercept and MR-PRESSO global tests confirmed that our MR analyses were not potentially affected by horizontal pleiotropy. These 40 immunophenotypes were also examined using MR Steiger directionality tests, which verified the directional stability of the immune signatures in patients with PCOS (Table S6).

Examination of Causal Links Between Immunophenotypes and SA Incidence Status

In this study, we identified notable causal effects involving 33 immune signatures among 731 immunophenotypes in individuals with SA (Figure 4). CCR2 (C-C chemokine receptor type 2) on CD14+ CD16+ monocyte, a possible protective factor, had the most significant causal relationship with SA (monocyte panel, OR: 0.975; 95% CI: 0.962–0.989; PIVW = 0.004). The CD28 and CD39 molecules in the Treg cell panel were also found to reduce the risk of developing SA, as did CD28+ DN (CD4−CD8−)%DN (Treg panel, OR: 0.974; 95% CI: 0.950–0.999; PIVW = 0.040), CD39+ CD8br%T cell (Treg panel, OR: 0.985; 95% CI: 0.971–0.999; PIVW = 0.036), and CD39+ CD8br%CD8br (Treg panel, OR: 0. 985; 95% CI: 0.972–0.999; PIVW = 0.036). In contrast, CD25 molecules on Treg cells increased the risk of developing SA, encompassing CD25++ CD8br%T cell (Treg panel, OR: 1.046; 95% CI: 1.008–1.085; PIVW = 0.017), CD28− CD25++ CD8br%T cell (Treg panel, OR: 1.072; 95% CI: 1.004–1.0145; PIVW = 0.039), and CD28− CD127− CD25++ CD8br%T cell (Treg panel, OR: 1.035; 95% CI: 1.007–1.064; PIVW = 0.014). Similarly, CD8 molecules on the TBNK panel, such as CD8br NKT%T cell (TBNK panel, OR: 1.039; 95% CI: 1.001–1.079; PIVW = 0.043) and CD8br NKT%lymphocyte (TBNK panel, OR: 1.045; 95% CI: 1.008–1.084; PIVW = 0.016), were also revealed as potential risk factors for SA. Intriguingly, the complicated patterns observed between the B-cell panel and SA population drew special attention, especially for CD25 molecules, which performed different roles in distinctive immune functions. For example, CD25 on IgD+ CD38br increased the risk of SA development (B-cell panel, OR: 1.039; 95% CI: 1.000–1.080; PIVW = 0.048), whereas CD25 on IgD− CD38dim had a protective effect on SA.

Figure 4 Causal associations between immune traits and SA: Results of MR analyses using the IVW method.

Abbreviations: SA, spontaneous abortion; MR, Mendelian randomization; IVW, inverse-variance weighted; OR, odds ratio.

The estimated analysis of the 33 causal interactions inferred from the IVW method received corroborative support from the MR‒Egger, weighted median, and MR-PRESSO approaches (Table S7). The significant protective impact of CCR2 on CD14+ CD16+ monocyte on SA was further established through additional assessments (monocyte panel, ORMR-Egger: 0.97, 95% CIMR-Egger: 0.96–0.99, PMR-Egger = 0.013; ORweighted median: 0.98, 95% CIweighted median: 0.96–1.00, Pweighted median = 0.032; ORMR-PRESSO: 0.98, 95% CIMR-PRESSO: 0.96–0.99, PMR-PRESSO = 0.0029) (Figure S3A and Table S7), and no significant outliers were found among IVs via LOO analysis (Figure S3B). According to the funnel plot, there was no indication of horizontal pleiotropy or heterogeneity, which further confirmed the credibility of the causal relationship between CCR2 on CD14+ CD16+ monocyte and SA (Figure S3C). Moreover, our investigation showed the absence of heterogeneity in the 33 causal effects, as evidenced by the results of Cochran’s Q statistic test (Table S8).

The MR‒Egger regression and MR-PRESSO global tests corroborated the absence of pleiotropy in our MR evaluations. The causal directions of the links between immune markers and SA were scrutinized and confirmed by MR-Steiger tests (Table S9).

Discussion

Utilizing a two-sample MR analysis approach, this study pioneers the comprehensive and impartial establishment of causal links between 731 immunophenotypes and reproductive health concerns, including AS, PCOS, and SA. Drawing on GWAS data at the gene level, this study investigated the intricate involvement of the immune system in the evolution of reproductive disorders. For AS, 34 causal links were established, notably, between the protective effects of the BAFF-R and CD27 molecules in the B-cell panel against AS. A pivotal causative link between CD11c+ CD62L− monocyte%monocyte (cDC panel) and AS pathogenesis was also identified. Regarding PCOS, our study confirmed 40 causal effects and underscored the vital roles of the CD20 and CD24 molecules in the B-cell panel in PCOS pathogenesis. CD4 on CM CD4+ (maturation stages of the T-cell panel) was found to significantly increase the risk of PCOS onset. Specifically, 33 causative associations were identified for SA, and the protective effect of CCR2 on CD14+ CD16+ monocyte (monocyte panel) on SA was elucidated. Our study also strongly emphasized the varied roles of the CD39, CD28, and CD25 molecules in the Treg cell panel in relation to SA. This study provides essential insights for further investigations of reproductive ill-health mechanisms, highlighting the pivotal role of the immune system in AS, PCOS, and SA.

An array of immune features significant for AS were identified in our study, particularly the protective causal relationship between BAFF-R and CD27 molecules in the B-cell panel. BAFF-R, a component of the tumor necrosis factor receptor (TNFR) family, is encoded by the TNFRSF13C gene located at chromosomal region 22q13. Its mRNA is translated into a transmembrane protein expressed on the surface of all Ig+ B cells.57,58 As a critical receptor for B-cell survival, BAFF-R may exert its protective effects against sperm abnormalities by regulating B-cell homeostasis in the testicular immune microenvironment. Previous studies have emphasized the essential role of BAFF-R signaling in maintaining the survival and function of mature B cells.59 Upon binding to BAFF, BAFF-R activates the PI3K/AKT signaling pathway, which is crucial for protein synthesis, metabolic adaptation, and cell survival.60 BAFF-R and its ligands also create independent homeostatic niches for B cell subsets,61,62 which are vital for maintaining B-cell homeostasis, particularly in complex immune environments like the testis. The specific mechanisms by which BAFF-R may lower the risk of AS should be explored in future experimental studies. Additionally, CD27, a key molecule guiding B cells toward plasma cell differentiation,63 has drawn attention for its potential role in reducing the risk of AS. Prior studies in chronic myelogenous leukemia have shown that CD27 interaction with CD70 activates Wnt target genes by enhancing the nuclear localization of active β-catenin and TNIK, promoting the proliferation and differentiation of leukemia stem cells.64 This suggests that CD27 may reduce the risk of AS by influencing similar processes in spermatogenesis, which involves the proliferation and differentiation of spermatogonia. Moreover, the mTOR signaling pathway plays a crucial role in regulating sperm quality, with mTOR activity suppressed in populations of highly viable spermatozoa.65 High CD27 expression has been reported to be closely linked to reduced mTOR activity.66 Therefore, it is speculated that CD27 may indirectly influence spermatogenesis by interacting with signaling pathways such as mTOR, affecting processes like cell proliferation, differentiation, and metabolism. Given the critical roles of BAFF-R and CD27 in B-cell function, the lack of extensive research on their roles in AS underscores the urgent need for further studies on these molecules as a potential protective factor in AS therapy.

PCOS is characterized by a multifaceted pathophysiology, with chronic low-grade inflammation being predominant, intertwining immune and metabolic imbalances.67 This study investigated the pathogenesis of PCOS from a causative perspective, focusing particularly on the complex role of CD4 on CM CD4+ (maturation stages of the T-cell panel), as well as CD20 and CD24 on the B-cell panel, in increasing the risk of PCOS. Previous studies reported a significantly higher frequency of CD4(+) CD28(null) T cells in PCOS patients.68 This study was the first to identify a causal link between CD4 on CM CD4+ T cells and an increased risk of PCOS. Abnormal activation of CD4+ T cells and alterations in the number of central memory T cells (TCM) have been documented to be associated with the development of several immune-related diseases.69,70 Given the immune dysregulation and chronic inflammation in PCOS, along with the complex roles of CD4+ TCM cells in immune regulation,70–72 it is suggested that CD4+ TCM may increase PCOS risk by disrupting immune balance, thereby affecting ovarian function and endocrine metabolism. In addition to T-cell dysfunction, B-cell abnormalities also play a crucial role in immune regulation in PCOS. CD20, due to its aberrant high expression in many diseases, has become an important therapeutic target in various conditions, such as B-cell lymphomas. CD20 induces cytosolic calcium flux through interactions with the B-cell receptor and activates related signaling pathways. Its dysregulation is closely associated with immune-related diseases.73–76 This suggests that CD20 on B cells may influence the inflammatory microenvironment of PCOS through similar mechanisms, by regulating inflammatory signaling pathways and affecting the production and release of inflammatory factors, thereby contributing to the pathological progression of PCOS. Moreover, CD24 expression is closely linked to B-cell energy metabolism and intracellular signaling pathways,77 and can enhance DNA damage-induced apoptosis by regulating the NF-κB signaling pathway.78 CD24 has been identified as a potential biomarker in diseases such as ovarian cancer, where it is associated with disease development, invasion, and metastasis.79 Changes in CD24 expression are closely related to ovulation, as it participates in regulating prostaglandin synthesis and transport, making it a target for the treatment of ovulatory disorders.80 This indicates that CD24 may play a similar role in the pathological process of PCOS, and further investigation is needed to determine its precise function in this context.

The aetiology of SA may be closely associated with abnormal inflammation.81 This study highlighted several key immune factors that influence the risk of SA, including CD39, CD28, CD25, and CCR2. Notably, the protective association of CD39 with SA revealed in this study demonstrates conserved characteristics across different species. Mouse model studies showed that CD39 activity was crucial for preventing miscarriage in antiphospholipid syndrome (APS)-induced abortion models.82 In CD39-overexpressing mice, abortion induced by antiphospholipid antibodies (aPL-ab) was prevented, and there was a reduction in trophoblast TF expression, C3d deposition, lipid peroxidation, and TNF-α expression. These findings suggested that CD39 might prevent miscarriage by regulating inflammatory and coagulation-related mechanisms. These findings are in agreement with human data, further confirming the protective role of CD39. Although the GWAS data in our study were derived from a European population, cross-population comparisons revealed similar findings in Asian populations, further indicating the key role of CD39 in maintaining immune balance at the maternal-fetal interface and preventing miscarriage.83 Recent studies on recurrent SA in Asian populations show reduced CD39+ cells in the decidua, possibly due to TGF-β-mTOR-HIF-1α pathway downregulation. The conserved nature of these findings across species and populations suggests that CD39-mediated immune regulation may be an evolutionarily conserved mechanism for maintaining pregnancy in mammals, further reinforcing the universal significance and relevance of our MR study.

In addition to CD39, CD28 expressed on Treg also plays critical roles in regulating immune responses, which influences the risk of SA. CD28, a homodimeric stimulatory cell surface receptor of the Ig superfamily,84 is essential for the activation of helper T cells, and its persistence is required for helper T cell polarization in response to infection.85 Prior studies have demonstrated that the second co-stimulatory signal provided by CD28 in the matrix plays a significant role in the embryo implantation process in mice.86 This suggests that CD28 might also be involved in pregnancy-related processes, which is consistent with our findings indicating its role in reducing the risk of SA. In contrast, high expression of CD25 on CD8br Treg cells was disclosed to be significantly associated with an increased risk of SA. Overexpression of CD25, the alpha chain of the IL-2 receptor, increases the receptor’s affinity for IL-2, which may lead to excessive consumption of IL-2.87 This reduction in local IL-2 concentrations can impair the activation and function of other immune cells, such as CD8+ and CD4+ T cells, even causing their functional exhaustion.88,89 Meanwhile, high CD25 expression may enhance IL-2 signaling, increasing pro-inflammatory cytokine secretion such as interferon-gamma, thereby triggering an inflammatory response.90 These immune alterations, collectively, may contribute to the increased risk of SA. Furthermore, this study provides the first evidence of a protective causal association between CCR2 on CD14+ CD16+ monocytes and SA. Earlier research has demonstrated that CD16+ monocytes play a crucial role in steady-state immune surveillance,91 and that CCR2+ monocytes are significantly involved in the repair of cerebrovascular damage caused by chronic social defeat stress.92 These findings suggest that CCR2 on CD14+ CD16+monocytes may reduce the risk of SA by modulating immune homeostasis and tissue repair processes.

This study performed elaborate MR analyses of 731 immunophenotypes, drawn from the latest GWAS cohort data. We uncovered causative links between immune traits and several reproductive ill-health conditions, namely, AS, PCOS and SA, which opens new avenues for identifying the underlying immunogenic mechanisms in these reproductive disorders. Nonetheless, there are certain limitations to our study. First, the significant causal relationships identified require further experimental validation. Second, the absence of available detailed personal data hindered a further stratified analysis of the population. Finally, as this study relied on European data, our findings cannot be applied to other ethnicities, thus constraining the generalizability of our results.

Conclusion

In summary, our study applied the MR approach to delineate the causal relationships between 731 immunophenotypes and reproductive health concerns, including AS, PCOS, and SA. We elucidated the intricate interplay between the immune system and reproductive diseases and filled a crucial void in related fields, with a focus on specific immunophenotypes. These findings expand the understanding of immune mechanisms and provide valuable insights for targeted prevention, early diagnosis, and the development of tailored therapeutic strategies for reproductive health issues.

Data Sharing Statement

Publicly available datasets were analyzed in this study. Summary statistics for abnormal spermatozoa were obtained from the FinnGen consortium R7 release data (https://r7.finngen.fi/). Statistics on polycystic ovary syndrome and spontaneous abortion were obtained from the FinnGen consortium R9 release data (https://r9.finngen.fi/). GWAS summary statistics for 731 immune traits could be publicly available in the GWAS Catalog (https://www.ebi.ac.uk/gwas/, accession numbers from GCST0001391 to GCST0002121).

Ethics Approval Statement

This study is based on publicly available, de-identified summary-level genetic data obtained from previous genome-wide association studies (GWAS), in which ethical approval and informed consent were obtained by the original authors. According to item 1 and 2 of Article 32 of the Measures for Ethical Review of Life Science and Medical Research Involving Human Subjects (dated February 18, 2023, China), our study does not involve direct interaction with human participants, biological samples, or identifiable personal data; thus, it is exempt from ethical review by our Institutional Review Board (IRB).

Consent for Publication

All authors read and approved the final version of the manuscript for publication.

Acknowledgments

Shuang Chen, Shihao Sun, Zihan Zhou are co-senior authors for this study. We extend our gratitude to the participants and researchers of the FinnGen and GWAS Catalog projects for their generous public sharing of GWAS summary data, which has greatly facilitated the conduct of this research.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This study was supported by the National Natural Science Foundation of China (grant number 82471677) and the Zhongyuan Medical and Health Leading Talent Project under the Zhongyuan Talent Program.

Disclosure

The authors declare that they have no competing interests in this work.

References

1. Zegers-Hochschild F, Adamson GD, Dyer S, et al. The international glossary on infertility and fertility care, 2017. Fertil Steril. 2017;108(3):393–406. doi:10.1016/j.fertnstert.2017.06.005

2. Dai M, Guo W, Zhu S, et al. Type 2 diabetes mellitus and the risk of abnormal spermatozoa: a Mendelian randomization study. Front Endocrinol. 2022;13:1035338. doi:10.3389/fendo.2022.1035338

3. Vollset SE, Goren E, Yuan CW, et al. Fertility, mortality, migration, and population scenarios for 195 countries and territories from 2017 to 2100: a forecasting analysis for the global burden of disease study. Lancet. 2020;396(10258):1285–1306. doi:10.1016/S0140-6736(20)30677-2

4. Zhang M, Liu X, Xu X, et al. The reference value of anti-Müllerian hormone to diagnose polycystic ovary syndrome is inversely associated with BMI: a retrospective study. Reprod Biol Endocrinol. 2023;21(1):15. doi:10.1186/s12958-023-01064-y

5. Elshazzly M, Lopez MJ, Reddy V, Embryology CO. Central Nervous System. Treasure Island (FL) ineligible companies: StatPearls; 2023. Disclosure: Michael Lopez declares no relevant financial relationships with ineligible companies. Disclosure: Vamsi Reddy declares no relevant financial relationships with ineligible companies. Disclosure: Omar Caban declares no relevant financial relationships with ineligible companies.: StatPearls PublishingCopyright © 2023, StatPearls Publishing LLC.

6. Kumar N, Singh AK. Trends of male factor infertility, an important cause of infertility: a review of literature. J Hum Reprod Sci. 2015;8(4):191–196. doi:10.4103/0974-1208.170370

7. Gill K, Jakubik J, Rosiak-Gill A, et al. Utility and predictive value of human standard semen parameters and sperm DNA dispersion for fertility potential. Int J Environ Res Public Health. 2019;16(11):2004. doi:10.3390/ijerph16112004

8. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet. 2007;370(9588):685–697. doi:10.1016/S0140-6736(07)61345-2

9. March WA, Moore VM, Willson KJ, Phillips DI, Norman RJ, Davies MJ. The prevalence of polycystic ovary syndrome in a community sample assessed under contrasting diagnostic criteria. Hum Reprod. 2010;25(2):544–551. doi:10.1093/humrep/dep399

10. Goodarzi MO, Dumesic DA, Chazenbalk G, Azziz R. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat Rev Endocrinol. 2011;7(4):219–231. doi:10.1038/nrendo.2010.217

11. Chen ZJ, Zhao H, He L, et al. Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat Genet. 2011;43(1):55–59. doi:10.1038/ng.732

12. Kandaraki E, Christakou C, Diamanti-Kandarakis E. Metabolic syndrome and polycystic ovary syndrome. and vice versa. Arq Bras Endocrinol Metabol. 2009;53(2):227–237. doi:10.1590/S0004-27302009000200014

13. Almeida ND, Basso O, Abrahamowicz M, Gagnon R, Tamblyn R. Risk of miscarriage in women receiving antidepressants in early pregnancy, correcting for induced abortions. Epidemiology. 2016;27(4):538–546. doi:10.1097/EDE.0000000000000484

14. Hertz-Picciotto I, Samuels SJ. Incidence of early loss of pregnancy. N Engl J Med. 1988;319(22):1483–1484.

15. Sapra KJ, Buck Louis GM, Sundaram R, et al. Signs and symptoms associated with early pregnancy loss: findings from a population-based preconception cohort. Hum Reprod. 2016;31(4):887–896. doi:10.1093/humrep/dew010

16. Lessey BA, Young SL. What exactly is endometrial receptivity? Fertil Steril. 2019;111(4):611–617. doi:10.1016/j.fertnstert.2019.02.009

17. Larsen EC, Christiansen OB, Kolte AM, Macklon N. New insights into mechanisms behind miscarriage. BMC Med. 2013;11:154. doi:10.1186/1741-7015-11-154

18. Parvanov D, Ganeva R, Vidolova N, Stamenov G. Decreased number of p16-positive senescent cells in human endometrium as a marker of miscarriage. J Assist Reprod Genet. 2021;38(8):2087–2095. doi:10.1007/s10815-021-02182-5

19. Zhou Y, Ding X, Wei H. Reproductive immune microenvironment. J Reprod Immunol. 2022;152:103654. doi:10.1016/j.jri.2022.103654

20. Meinhardt A, Dejucq-Rainsford N, Bhushan S. Testicular macrophages: development and function in health and disease. Trends Immunol. 2022;43(1):51–62. doi:10.1016/j.it.2021.11.003

21. Scott JR. Reproductive immunology from the perspective of the clinician. J Reprod Immunol. 2019;133:27–29. doi:10.1016/j.jri.2019.05.002

22. Yang X, Gilman-Sachs A, Kwak-Kim J. Ovarian and endometrial immunity during the ovarian cycle. J Reprod Immunol. 2019;133:7–14. doi:10.1016/j.jri.2019.04.001

23. Arck P, Solano ME, Walecki M, Meinhardt A. The immune privilege of testis and gravid uterus: same difference? Mol Cell Endocrinol. 2014;382(1):509–520. doi:10.1016/j.mce.2013.09.022

24. Bellgrau D, Gold D, Selawry H, Moore J, Franzusoff A, Duke RC. A role for CD95 ligand in preventing graft rejection. Nature. 1995;377(6550):630–632. doi:10.1038/377630a0

25. Niederkorn JY. See no evil, hear no evil, do no evil: the lessons of immune privilege. Nat Immunol. 2006;7(4):354–359. doi:10.1038/ni1328

26. Zhao S, Zhu W, Xue S, Han D. Testicular defense systems: immune privilege and innate immunity. Cell Mol Immunol. 2014;11(5):428–437. doi:10.1038/cmi.2014.38

27. Lokka E, Lintukorpi L, Cisneros-Montalvo S, et al. Generation, localization and functions of macrophages during the development of testis. Nat Commun. 2020;11(1):4375. doi:10.1038/s41467-020-18206-0

28. Wang M, Fijak M, Hossain H, et al. Characterization of the micro-environment of the testis that shapes the phenotype and function of testicular macrophages. J Immunol. 2017;198(11):4327–4340. doi:10.4049/jimmunol.1700162

29. Bukovsky A, Presl J. Role of the immune system in regulation of ovarian function-hypothesis. Czech Med. 1978;1(4):229–237.

30. Kinnear HM, Tomaszewski CE, Chang FL, et al. The ovarian stroma as a new frontier. Reproduction. 2020;160(3):R25–r39. doi:10.1530/REP-19-0501

31. Norman RJ, Brannstrom M. White cells and the ovary--incidental invaders or essential effectors? J Endocrinol. 1994;140(3):333–336. doi:10.1677/joe.0.1400333

32. Robertson SA, Moldenhauer LM, Green ES, Care AS, Hull ML. Immune determinants of endometrial receptivity: a biological perspective. Fertil Steril. 2022;117(6):1107–1120. doi:10.1016/j.fertnstert.2022.04.023

33. Lessey BA. Assessment of endometrial receptivity. Fertil Steril. 2011;96(3):522–529. doi:10.1016/j.fertnstert.2011.07.1095

34. Aplin JD, Ruane PT. Embryo-epithelium interactions during implantation at a glance. J Cell Sci. 2017;130(1):15–22. doi:10.1242/jcs.175943

35. Hiby SE, Apps R, Sharkey AM, et al. Maternal activating KIRs protect against human reproductive failure mediated by fetal HLA-C2. J Clin Invest. 2010;120(11):4102–4110. doi:10.1172/JCI43998

36. Smith GD, Ebrahim S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? Int J Epidemiol. 2003;32(1):1–22. doi:10.1093/ije/dyg070

37. Sekula P, Del Greco MF, Pattaro C, Köttgen A. Mendelian randomization as an approach to assess causality using observational data. J Am Soc Nephrol. 2016;27(11):3253–3265. doi:10.1681/ASN.2016010098

38. Hemani G, Zheng J, Elsworth B, et al. The MR-base platform supports systematic causal inference across the human phenome. Elife. 2018;7:e34408.

39. Boef AG, Dekkers OM, le Cessie S. Mendelian randomization studies: a review of the approaches used and the quality of reporting. Int J Epidemiol. 2015;44(2):496–511. doi:10.1093/ije/dyv071

40. Spiga F, Gibson M, Dawson S, et al. Tools for assessing quality and risk of bias in Mendelian randomization studies: a systematic review. Int J Epidemiol. 2023;52(1):227–249. doi:10.1093/ije/dyac149

41. Minelli C, Thompson JR, Tobin MD, Abrams KR. An integrated approach to the meta-analysis of genetic association studies using Mendelian randomization. Am J Epidemiol. 2004;160(5):445–452. doi:10.1093/aje/kwh228

42. Choi KW, Chen CY, Stein MB, et al. Assessment of bidirectional relationships between physical activity and depression among adults: a 2-sample mendelian randomization study. JAMA Psychiatry. 2019;76(4):399–408. doi:10.1001/jamapsychiatry.2018.4175

43. Georgakis MK, Gill D. Mendelian randomization studies in stroke: exploration of risk factors and drug targets with human genetic data. Stroke. 2021;52(9):2992–3003. doi:10.1161/STROKEAHA.120.032617

44. Kennedy OJ, Pirastu N, Poole R, et al. Coffee consumption and kidney function: a Mendelian randomization study. Am J Kidney Dis. 2020;75(5):753–761. doi:10.1053/j.ajkd.2019.08.025

45. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ. 2018;362:k601. doi:10.1136/bmj.k601

46. Orrù V, Steri M, Sidore C, et al. Complex genetic signatures in immune cells underlie autoimmunity and inform therapy. Nat Genet. 2020;52(10):1036–1045. doi:10.1038/s41588-020-0684-4

47. Auton A, Brooks LD, Durbin RM, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68–74. doi:10.1038/nature15393

48. Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet Epidemiol. 2013;37(7):658–665. doi:10.1002/gepi.21758

49. Pierce BL, Ahsan H, Vanderweele TJ. Power and instrument strength requirements for Mendelian randomization studies using multiple genetic variants. Int J Epidemiol. 2011;40(3):740–752. doi:10.1093/ije/dyq151

50. Burgess S, Small DS, Thompson SG. A review of instrumental variable estimators for Mendelian randomization. Stat Methods Med Res. 2017;26(5):2333–2355. doi:10.1177/0962280215597579

51. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–314. doi:10.1002/gepi.21965

52. Burgess S, Thompson SG. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32(5):377–389. doi:10.1007/s10654-017-0255-x

53. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50(5):693–698. doi:10.1038/s41588-018-0099-7

54. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327(7414):557–560. doi:10.1136/bmj.327.7414.557

55. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–525. doi:10.1093/ije/dyv080

56. Hemani G, Tilling K, Davey Smith G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 2017;13(11):e1007081. doi:10.1371/journal.pgen.1007081

57. Mackay F, Schneider P, Rennert P, Browning J. BAFF AND April: a tutorial on B cell survival. Annu Rev Immunol. 2003;21:231–264. doi:10.1146/annurev.immunol.21.120601.141152

58. Ng LG, Sutherland AP, Newton R, et al. B cell-activating factor belonging to the TNF family (BAFF)-R is the principal BAFF receptor facilitating BAFF costimulation of circulating T and B cells. J Immunol. 2004;173(2):807–817. doi:10.4049/jimmunol.173.2.807

59. Rauch M, Tussiwand R, Bosco N, Rolink AG. Crucial role for BAFF-BAFF-R signaling in the survival and maintenance of mature B cells. PLoS One. 2009;4(5):e5456. doi:10.1371/journal.pone.0005456

60. Sevdali E, Block V, Lataretu M, et al. BAFFR activates PI3K/AKT signaling in human naive but not in switched memory B cells through direct interactions with B cell antigen receptors. Cell Rep. 2022;39(13):111019. doi:10.1016/j.celrep.2022.111019

61. Naradikian MS, Perate AR, Cancro MP. BAFF receptors and ligands create independent homeostatic niches for B cell subsets. Curr Opin Immunol. 2015;34:126–129. doi:10.1016/j.coi.2015.03.005

62. Wensveen FM, Slinger E, van Attekum MH, Brink R, Eldering E. Antigen-affinity controls pre-germinal center B cell selection by promoting Mcl-1 induction through BAFF receptor signaling. Sci Rep. 2016;6:35673. doi:10.1038/srep35673

63. Agematsu K, Hokibara S, Nagumo H, Shinozaki K, Yamada S, Komiyama A. Plasma cell generation from B-lymphocytes via CD27/CD70 interaction. Leuk Lymphoma. 1999;35(3–4):219–225. doi:10.3109/10428199909145724

64. Schürch C, Riether C, Matter MS, Tzankov A, Ochsenbein AF. CD27 signaling on chronic myelogenous leukemia stem cells activates Wnt target genes and promotes disease progression. J Clin Invest. 2012;122(2):624–638. doi:10.1172/JCI45977

65. Silva JV, Cabral M, Correia BR, et al. mTOR signaling pathway regulates sperm quality in older men. Cells. 2019;8(6). doi:10.3390/cells8060629.

66. Ansari AW, Jayakumar MN, Ahmad F, et al. Azithromycin targets the CD27 pathway to modulate CD27hi T-lymphocyte expansion and type-1 effector phenotype. Front Immunol. 2024;15:1447625. doi:10.3389/fimmu.2024.1447625

67. Wang J, Yin T, Liu S. Dysregulation of immune response in PCOS organ system. Front Immunol. 2023;14:1169232. doi:10.3389/fimmu.2023.1169232

68. Niccoli G, Apa R, Lanzone A, et al. CD4+CD28 null T lymphocytes are expanded in young women with polycystic ovary syndrome. Fertil Steril. 2011;95(8):2651–2654. doi:10.1016/j.fertnstert.2011.01.129

69. Zhao W, Dong Y, Wu C, Ma Y, Ji Y, Jin Y. TIGIT overexpression diminishes the function of CD4 T cells and ameliorates the severity of rheumatoid arthritis in mouse models. Exp Cell Res. 2016;340(1):132–138. doi:10.1016/j.yexcr.2015.12.002

70. Yu J, Long B, Li Z, et al. Central memory CD4+ T cells play a protective role against immune checkpoint inhibitor-associated myocarditis. Cardiovasc Res. 2024;120(12):1442–1455. doi:10.1093/cvr/cvae133

71. Zhang X, Xiao X, Chang Li X, Sun R, Tian Z, Wei H. CD4(+)CD62L(+) central memory T cells can be converted to Foxp3(+) T cells. PLoS One. 2013;8(10):e77322. doi:10.1371/journal.pone.0077322

72. Haqqani AA, Marek SL, Kumar J, Davenport M, Wang H, Tilton JC. Central memory CD4+ T cells are preferential targets of double infection by HIV-1. Virol J. 2015;12:184. doi:10.1186/s12985-015-0415-0

73. Walshe CA, Beers SA, French RR, et al. Induction of cytosolic calcium flux by CD20 is dependent upon B Cell antigen receptor signaling. J Biol Chem. 2008;283(25):16971–16984. doi:10.1074/jbc.M708459200

74. Polyak MJ, Li H, Shariat N, Deans JP. CD20 homo-oligomers physically associate with the B cell antigen receptor. Dissociation upon receptor engagement and recruitment of phosphoproteins and calmodulin-binding proteins. J Biol Chem. 2008;283(27):18545–18552. doi:10.1074/jbc.M800784200

75. van de Ven AA, Compeer EB, Bloem AC, et al. Defective calcium signaling and disrupted CD20-B-cell receptor dissociation in patients with common variable immunodeficiency disorders. J Allergy Clin Immunol. 2012;129(3):755–761.e757. doi:10.1016/j.jaci.2011.10.020

76. Pavlasova G, Mraz M. The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy. Haematologica. 2020;105(6):1494–1506. doi:10.3324/haematol.2019.243543

77. Mensah FFK, Armstrong CW, Reddy V, et al. CD24 expression and B cell maturation shows a novel link with energy metabolism: potential implications for patients with myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol. 2018;9:2421. doi:10.3389/fimmu.2018.02421

78. Ju JH, Jang K, Lee KM, et al. CD24 enhances DNA damage-induced apoptosis by modulating NF-κB signaling in CD44-expressing breast cancer cells. Carcinogenesis. 2011;32(10):1474–1483. doi:10.1093/carcin/bgr173

79. Tarhriz V, Bandehpour M, Dastmalchi S, Ouladsahebmadarek E, Zarredar H, Eyvazi S. Overview of CD24 as a new molecular marker in ovarian cancer. J Cell Physiol. 2019;234(3):2134–2142. doi:10.1002/jcp.27581

80. Dong JP, Dai ZH, Jiang ZX, et al. CD24: a marker of granulosa cell subpopulation and a mediator of ovulation. Cell Death Dis. 2019;10(11):791. doi:10.1038/s41419-019-1995-1

81. Gao P, Zha Y, Gong X, Qiao F, Liu H. The role of maternal-foetal interface inflammation mediated by NLRP3 inflammasome in the pathogenesis of recurrent spontaneous abortion. Placenta. 2020;101:221–229. doi:10.1016/j.placenta.2020.09.067

82. Samudra AN, Dwyer KM, Selan C, et al. CD39 and CD73 activity are protective in a mouse model of antiphospholipid antibody-induced miscarriages. J Autoimmun. 2018;88:131–138. doi:10.1016/j.jaut.2017.10.009

83. Zhu J, Song G, Zhou X, et al. CD39/CD73 dysregulation of adenosine metabolism increases decidual natural killer cell cytotoxicity: implications in unexplained recurrent spontaneous abortion. Front Immunol. 2022;13:813218. doi:10.3389/fimmu.2022.813218

84. Beyersdorf N, Kerkau T, Hünig T. CD28 co-stimulation in T-cell homeostasis: a recent perspective. Immunotargets Ther. 2015;4:111–122. doi:10.2147/ITT.S61647

85. Linterman MA, Denton AE, Divekar DP, et al. CD28 expression is required after T cell priming for helper T cell responses and protective immunity to infection. Elife. 2014;3:e03180.

86. Liu S, He J, Chen X, et al. Costimulatory molecule CD28 participates in the process of embryo implantation in mice. Reprod Sci. 2014;21(6):686–695. doi:10.1177/1933719113512537

87. Wang X, Rickert M, Garcia KC. Structure of the quaternary complex of interleukin-2 with its alpha, beta, and gammac receptors. Science. 2005;310(5751):1159–1163. doi:10.1126/science.1117893

88. Balkhi MY, Ma Q, Ahmad S, Junghans RP. T cell exhaustion and Interleukin 2 downregulation. Cytokine. 2015;71(2):339–347. doi:10.1016/j.cyto.2014.11.024

89. Kastenmuller W, Gasteiger G, Subramanian N, et al. Regulatory T cells selectively control CD8+ T cell effector pool size via IL-2 restriction. J Immunol. 2011;187(6):3186–3197. doi:10.4049/jimmunol.1101649

90. Joosse ME, Charbit-Henrion F, Boisgard R, et al. Duplication of the IL2RA locus causes excessive IL-2 signaling and may predispose to very early onset colitis. Mucosal Immunol. 2021;14(5):1172–1182. doi:10.1038/s41385-021-00423-5

91. Waschbisch A, Schröder S, Schraudner D, et al. Pivotal role for CD16+ monocytes in immune surveillance of the central nervous system. J Immunol. 2016;196(4):1558–1567. doi:10.4049/jimmunol.1501960

92. Lehmann ML, Samuels JD, Kigar SL, Poffenberger CN, Lotstein ML, Herkenham M. CCR2 monocytes repair cerebrovascular damage caused by chronic social defeat stress. Brain Behav Immun. 2022;101:346–358. doi:10.1016/j.bbi.2022.01.011

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.