Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.006
Filter
1.
PLoS One ; 19(6): e0304591, 2024.
Article in English | MEDLINE | ID: mdl-38843259

ABSTRACT

OBJECTIVE: The systemic immuno-inflammatory index (SII), a novel immune marker of inflammation, has not been previously associated with endometriosis. The objective of this research is to explore the link between SII and the occurrence of endometriosis. METHODS: Utilizing data from the National Health and Nutrition Examination Survey (NHANES) spanning 2001 to 2006, we screened and extracted relevant information from the population. Participants missing data on either SII or endometriosis were excluded. We divided the remaining cohort into quartiles based on SII levels: Q1 (SII < 249, n = 848), Q2 (249 ≤ SII < 604.55, n = 847), Q3 (604.55 ≤ SII < 825.35, n = 847), and Q4 (SII ≥ 852.35, n = 848). Multiple linear regression and smooth curve fitting techniques, were to evaluate the non-linear association between SII and endometriosis. RESULTS: The study included 3,390 adults aged 20 to 55. Multiple linear regression analysis revealed a significant positive correlation between SII and endometriosis [3.14, 95% CI (2.22, 4.45), P < 0.01]. This correlation was consistent across subgroups defined by marital status, poverty income ratio, BMI, alcohol consumption, and age at first menstrual period. However, the relationship between SII and endometriosis was significantly modified by age, education, and history of pregnancy in the stratified analyses. The curve fitting indicated an S-shaped curve, with an inflection point at SII = 1105.76. CONCLUSION: The SII may serve as a predictive marker for endometriosis risk among women in the United States, offering a potentially simple and cost-effective approach. However, given the cross-sectional design of this investigation, further validation in prospective studies is necessary.


Subject(s)
Biomarkers , Endometriosis , Inflammation , Nutrition Surveys , Humans , Endometriosis/immunology , Endometriosis/epidemiology , Female , Adult , Cross-Sectional Studies , Middle Aged , Young Adult , United States/epidemiology
2.
Int J Mol Sci ; 25(9)2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38732021

ABSTRACT

The most common manifestation of endometriosis, a condition characterized by the presence of endometrial-like tissue outside of the uterus, is the endometrioma, a cystic ovarian lesion. It is a commonly occurring condition associated with chronic pelvic pain exacerbated prior to and during menstruation, as well as infertility. The exact pathomechanisms of the endometrioma are still not fully understood. Emerging evidence suggests a pivotal role of immune dysregulation in the pathogenesis of endometriomas, primarily influencing both local and systemic inflammatory processes. Among the factors implicated in the creation of the inflammatory milieu associated with endometriomas, alterations in both serum and local levels of several cytokines stand out, including IL-6, IL-8, and IL-1ß, along with abnormalities in the innate immune system. While numerous signaling pathways have been suggested to play a role in the inflammatory process linked to endometriomas, only NF-κB has been conclusively demonstrated to be involved. Additionally, increased oxidative stress, both resulting from and contributing to endometriomas, has been identified as a primary driver of both systemic and local inflammation associated with the condition. This article reviews the current understanding of immune dysfunctions in the endometrioma and their implications for inflammation.


Subject(s)
Endometriosis , Inflammation , Humans , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/metabolism , Female , Inflammation/immunology , Inflammation/pathology , Cytokines/metabolism , Oxidative Stress , Signal Transduction , Immunity, Innate , Animals
3.
Clinics (Sao Paulo) ; 79: 100390, 2024.
Article in English | MEDLINE | ID: mdl-38781760

ABSTRACT

Endometriosis's pathophysiology remains incompletely understood, with evidence pointing towards a dysregulated immune response. Regulatory T (Treg) cells, pivotal in maintaining self-tolerance, may facilitate the survival of ectopic endometrial cells within the abdominal cavity, thereby contributing to endometriosis development. This study aimed to assess the prevalence of CD39+CD73+ suppressor Treg cell subsets in the peripheral blood of endometriosis patients. This research focuses on the pivotal role of regulatory T-cells (Tregs), which are essential for maintaining immune tolerance and preventing autoimmune diseases. A case-control study was conducted, including 32 women diagnosed with endometriosis and 22 control subjects. The frequency of peripheral blood CD39+CD73+ suppressor Treg cells was quantified using flow cytometry. No significant differences were observed in the frequency of CD3+CD4+CD25High cells (Median [M]: 10.1; Interquartile Range [IQR]: 6.32‒18.3 vs. M: 9.72; IQR: 6.22-19.8) or CD3+CD4+CD25HighCD39+Foxp3+ cells (M: 31.1; IQR: 19.7-44.0 vs. M: 30.55; IQR: 18.5-45.5) between controls and patients. However, a significantly lower frequency of CD3+CD4+CD25HighCD39+CD73+ cells was observed in the endometriosis group compared to controls (M: 1.98; IQR: 0.0377-3.17 vs. M: 2.25; IQR: 0.50-4.08; p = 0.0483), suggesting a reduction in systemic immune tolerance among these patients. This finding highlights the potential role of CD39 and CD73 expression on Treg cells as biomarkers for assessing disease severity and progression. Furthermore, elucidating the mechanisms driving these alterations may unveil new therapeutic strategies to restore immune equilibrium and mitigate endometriosis symptoms.


Subject(s)
Apyrase , Endometriosis , Flow Cytometry , Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Humans , Female , Endometriosis/immunology , Endometriosis/blood , T-Lymphocytes, Regulatory/immunology , Adult , Case-Control Studies , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/analysis , Apyrase/analysis , 5'-Nucleotidase/blood , Young Adult , Antigens, CD/blood , Antigens, CD/analysis , Statistics, Nonparametric , Reference Values
4.
Front Immunol ; 15: 1339647, 2024.
Article in English | MEDLINE | ID: mdl-38660311

ABSTRACT

Introduction: Over the past decades, immune dysregulation has been consistently demonstrated being common charactoristics of endometriosis (EM) and Inflammatory Bowel Disease (IBD) in numerous studies. However, the underlying pathological mechanisms remain unknown. In this study, bioinformatics techniques were used to screen large-scale gene expression data for plausible correlations at the molecular level in order to identify common pathogenic pathways between EM and IBD. Methods: Based on the EM transcriptomic datasets GSE7305 and GSE23339, as well as the IBD transcriptomic datasets GSE87466 and GSE126124, differential gene analysis was performed using the limma package in the R environment. Co-expressed differentially expressed genes were identified, and a protein-protein interaction (PPI) network for the differentially expressed genes was constructed using the 11.5 version of the STRING database. The MCODE tool in Cytoscape facilitated filtering out protein interaction subnetworks. Key genes in the PPI network were identified through two topological analysis algorithms (MCC and Degree) from the CytoHubba plugin. Upset was used for visualization of these key genes. The diagnostic value of gene expression levels for these key genes was assessed using the Receiver Operating Characteristic (ROC) curve and Area Under the Curve (AUC) The CIBERSORT algorithm determined the infiltration status of 22 immune cell subtypes, exploring differences between EM and IBD patients in both control and disease groups. Finally, different gene expression trends shared by EM and IBD were input into CMap to identify small molecule compounds with potential therapeutic effects. Results: 113 differentially expressed genes (DEGs) that were co-expressed in EM and IBD have been identified, comprising 28 down-regulated genes and 86 up-regulated genes. The co-expression differential gene of EM and IBD in the functional enrichment analyses focused on immune response activation, circulating immunoglobulin-mediated humoral immune response and humoral immune response. Five hub genes (SERPING1、VCAM1、CLU、C3、CD55) were identified through the Protein-protein Interaction network and MCODE.High Area Under the Curve (AUC) values of Receiver Operating Characteristic (ROC) curves for 5hub genes indicate the predictive ability for disease occurrence.These hub genes could be used as potential biomarkers for the development of EM and IBD. Furthermore, the CMap database identified a total of 9 small molecule compounds (TTNPB、CAY-10577、PD-0325901 etc.) targeting therapeutic genes for EM and IBD. Discussion: Our research revealed common pathogenic mechanisms between EM and IBD, particularly emphasizing immune regulation and cell signalling, indicating the significance of immune factors in the occurence and progression of both diseases. By elucidating shared mechanisms, our study provides novel avenues for the prevention and treatment of EM and IBD.


Subject(s)
Endometriosis , Inflammatory Bowel Diseases , Protein Interaction Maps , Transcriptome , Humans , Endometriosis/immunology , Endometriosis/genetics , Female , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Computational Biology/methods , Gene Expression Profiling , Databases, Genetic , Gene Regulatory Networks , Biomarkers , Gene Expression Regulation
5.
Int Immunopharmacol ; 132: 111942, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38565045

ABSTRACT

Endometriosis (EM) is a gynecological inflammatory disease often accompanied by stress, chronic pelvic pain (CPP), anxiety, and depression, leading to a diminished quality of life. This review aims to discuss the relationship between systemic and local inflammatory responses in the central nervous system (CNS), focusing on glial dysfunctions (astrocytes and microglia) as in critical brain regions involved in emotion, cognition, pain processing, anxiety, and depression. The review presents that EM is connected to increased levels of pro-inflammatory cytokines in the circulation. Additionally, chronic stress and CPP as stressors may contribute to the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, depleting the production of inflammatory mediators in the circulatory system and the brain. The systemic cytokines cause blood-brain barrier (BBB) breakdown, activate microglia in the brain, and lead to neuroinflammation. Furthermore, CPP may induce neuronal morphological alterations in critical regions through central sensitization and the activation of glial cells. The activation of glial cells, particularly the polarization of microglia, leads to the activation of the NLRP3 inflammasome and the overproduction of inflammatory cytokines. These inflammatory cytokines interact with the signaling pathways involved in neural plasticity. Additionally, persistent inflammatory conditions in the brain lead to neuronal death, which is correlated with a reduced volume of key brain regions such as the hippocampus. This review highlights the involvement of glial cells in the pathogenesis of the mental comorbidities of EM (i.e., pain, anxiety, and depression) and to discuss potential therapeutic approaches for targeting the inflammation and activation of microglia in key brain regions.


Subject(s)
Anxiety , Depression , Endometriosis , Neuroglia , Humans , Female , Endometriosis/immunology , Endometriosis/pathology , Depression/immunology , Depression/etiology , Depression/metabolism , Anxiety/immunology , Animals , Neuroglia/immunology , Inflammation/immunology , Stress, Psychological/immunology , Cytokines/metabolism , Brain/immunology , Brain/pathology , Brain/metabolism
6.
J Reprod Immunol ; 163: 104242, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38503076

ABSTRACT

The innate and adaptive immune systems are the two key branches that determine host protection at all mucosal surfaces in human body, including the female reproductive tract. The pattern recognition receptors within the host that recognize pathogen-associated molecular patterns are expressed on the cells of the innate immune system. Rapidly reactive, theinnate immune system, responds immediately to the presence of infectious or other non-self agents, thereby launching an inflammatory response to protect the host until the activation of slower adaptive immune system. Macrophages, dendritic cells, and toll-like receptors are integral components of the innate immune system. In contrast, T-helper (Th1/Th2/Th17) cells and regulatory T (Treg) cells are the primary components of adaptive immune system. Studies showed that the growth and progression of endometriosis continue even in unilateral ovariectomized animal suggesting that besides ovarian steroid hormones, the growth of endometriosis could be regulated by innate/adaptive immune systems in pelvic environment. Recent reports demonstrated a potential role of Th1/Th2/Th17/Treg cells either individually or collectively in the initiation, maintenance, and progression of endometriosis. Herewe review the fundamental knowledge of innate and adaptive immunity and elaborate the role of innate and adaptive immunity in endometriosis based on both human and experimental data.


Subject(s)
Adaptive Immunity , Endometriosis , Immunity, Innate , Humans , Female , Endometriosis/immunology , Endometriosis/pathology , Animals , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/metabolism , Toll-Like Receptors/immunology
7.
Immunology ; 172(3): 469-485, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38544333

ABSTRACT

Endometriosis is defined as an oestrogen-dependent and inflammatory gynaecological disease of which the pathogenesis remains unclear. This study aimed to investigate the cellular heterogeneity and reveal the effect of CD8+ T cells on the progress of endometriosis. Three ovarian endometriosis patients were collected, and single-cell RNA sequencing (scRNA-seq) progressed and delineated the cellular landscape of endometriosis containing five cell clusters. The endometrial cells (EMCs) were the major component, of which the mesenchymal cells were preponderant and characterized with increased inflammation and oestrogen synthesis in endometriosis. The proportion of T cells, mainly CD8+ T cells rather than CD4+, was reduced in endometriotic lesions, and the cytokines and cytotoxicity of ectopic T cells were depressed. CD8+ T cells depressed the proliferation of ESCs through inhibiting CDK1/CCNB1 pathway to arrest the cell cycle and triggered inflammation through activating STAT1 pathway. Correspondingly, the coculture with ESCs resulted in the dysfunction of CD8+ T cells through upregulating STAT1/PDCD1 pathway and glycolysis-promoted metabolism reprogramming. The endometriotic lesions were larger in nude mouse models with T-cell deficiency than the normal mouse models. The inhibition of T cells via CD90.2 or CD8A antibody increased the endometriotic lesions in mouse models, and the supplement of T cells to nude mouse models diminished the lesion sizes. In conclusion, this study revealed the global cellular variation of endometriosis among which the cellular count and physiology of EMCs and T cells were significantly changed. The depressed cytotoxicity and aberrant metabolism of CD8+ T cells were induced by ESCs with the activation of STAT1/PDCD1 pathway resulting in immune survival to promote endometriosis.


Subject(s)
CD8-Positive T-Lymphocytes , Endometriosis , STAT1 Transcription Factor , Stromal Cells , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/metabolism , Female , CD8-Positive T-Lymphocytes/immunology , Humans , Animals , Mice , Stromal Cells/immunology , Stromal Cells/metabolism , STAT1 Transcription Factor/metabolism , Programmed Cell Death 1 Receptor/metabolism , Endometrium/immunology , Endometrium/pathology , Disease Models, Animal , Signal Transduction , Mice, Nude , Adult , CDC2 Protein Kinase/metabolism , Coculture Techniques , Cytokines/metabolism
8.
J Reprod Immunol ; 163: 104219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38422807

ABSTRACT

Endometriosis is a common and frequent disease in gynecology; its etiology and pathogenesis are partially understood and still not clear. The construction of suitable animal models is beneficial for basic research related to the disease. Currently, rodents have the advantages of low cost, fast reproduction, easy rearing, and a similar endometrial structure to humans. Depending on the purpose of the experiment, different molding methods have their advantages. In this paper, we describe the traditional methods of constructing endometriosis rodent models, compare their advantages and disadvantages, and introduce newly developed rodent models, such as cell line injection models, pain models, genetically engineered mouse models, fluorescent tracer models, iron overload models, chemical induction models, and methods of constructing rodent models of different subtypes of endometriosis. Fertility and treatment of endometriosis rodent models are also described. This study provides a reference for researchers in the selection of animal models for pathogenesis and drug treatment studies.


Subject(s)
Disease Models, Animal , Endometriosis , Endometriosis/pathology , Endometriosis/therapy , Endometriosis/immunology , Female , Animals , Humans , Mice , Rodentia , Endometrium/pathology , Rats
9.
Pol Merkur Lekarski ; 50(298): 232-236, 2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36086981

ABSTRACT

Endometriosis is an inflammatory estrogen-dependent gynecological disease characterized by the presence of endometrial tissue outside the uterine cavity. An important role in the pathogenesis of this disease is played by disorders of the immune system involving chemokines and their receptors, including the CXCL8-CXCR1/ 2 system. AIM: The aim of the study was to assess the concentration of the CXCL8 chemokine and its CXCR1 and CXCR2 receptors in the peritoneal fluid of women with endometriosis. MATERIALS AND METHODS: The study included 32 women aged 21 to 47 years with diagnosed endometriosis and a control group of 8 healthy women aged 21 to 40 years. The material for the research was the peritoneal fluid collected during the laparoscopic procedure. The concentration of chemokines was determined by ELISA tests. RESULTS: The conducted studies showed that the concentration of the CXCL8 chemokine was significantly higher in the peritoneal fluid of the studied women and depended on the clinical advancement of the disease. CONCLUSIONS: Changes in the concentration of the CXCL8 chemokine in the peritoneal fluid of women with endometriosis may indicate impaired immune response and indicate an inflammatory process within the peritoneal cavity. The demonstrated relationship between the concentration of CXCL8 and the stages of clinical advancement indicates a significant role of this chemokine in the development of the disease.


Subject(s)
Ascitic Fluid/chemistry , Endometriosis , Interleukin-8/analysis , Receptors, Interleukin-8A/analysis , Receptors, Interleukin-8B/analysis , Chemokines , Endometriosis/immunology , Female , Humans , Interleukin-8/physiology
11.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163463

ABSTRACT

Endometriosis, defined as the presence of endometrium-like tissue outside the uterus, is estimated to affect 10% of women of reproductive age [...].


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Endometriosis/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Drug Discovery , Endometriosis/drug therapy , Endometriosis/pathology , Female , Gene Expression Regulation/drug effects , Humans , Signal Transduction/drug effects
12.
Pharmacol Res Perspect ; 10(1): e00916, 2022 02.
Article in English | MEDLINE | ID: mdl-35084123

ABSTRACT

Endometriosis in an estrogen-dependent disease that is characterized by the presence of endometrial tissue outside the uterine cavity leading to pain and infertility in many affected women. Highly efficient treatment options which create a hypo-estrogenic environment can cause side effects such as hot flushes and bone mass loss that are not favorable for premenopausal women. Previous work has demonstrated that increased local or systemic prolactin seems to be involved in the pathogenesis of endometriosis. Here we examined two prolactin receptor (PRLR) blocking antibodies in a murine endometriosis interna model which relies on the induction of systemic hyperprolactinemia in female SHN mice. The severity of the disease is determined by the degree of endometrial invasion into the myometrium. In this model, endometriosis was inhibited by clinical gold standards such as progestins and anti-estrogenic approaches. PRLR blockade completely inhibited endometriosis in this mouse model to the same extent as the anti-estrogen faslodex or the GnRH antagonist cetrorelix. In contrast to cetrorelix and faslodex, the PRLR antibodies did not decrease relative uterine weights and were thus devoid of anti-estrogenic effects. We therefore hypothesize that PRLR antibodies may present a novel and highly efficient treatment option for endometriosis with a good safety and tolerability profile. Clinical studies are on the way to test this hypothesis.


Subject(s)
Antibodies/pharmacology , Endometriosis/therapy , Hormone Antagonists/pharmacology , Receptors, Prolactin/antagonists & inhibitors , Animals , Antibodies/toxicity , Disease Models, Animal , Endometriosis/immunology , Female , Fulvestrant/pharmacology , Fulvestrant/toxicity , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/toxicity , Hormone Antagonists/toxicity , Mice , Receptors, Prolactin/immunology
13.
J Reprod Immunol ; 149: 103453, 2022 02.
Article in English | MEDLINE | ID: mdl-34839179

ABSTRACT

Innate and adaptive immune factors play significant roles in the pathophysiology of endometriosis. T helper 17 (Th17) cells, a pro-inflammatory T cell subset, were considered to contribute to the progression of endometriosis lesions. However, the regulatory mechanisms of Th17 cells in endometriosis remain unidentified, partially due to the difficulty in recovering live Th17 cells from endometriosis patients. In this study, by flow cytometry analysis of a set of chemokine receptors including CXCR3, CCR4, CCR10, and CCR6, live RORγt-and-IL-17A-expressing Th17 cells were enriched from peritoneal fluid (PF) of patients with different stages of endometriosis for the first time, RNA-sequencing (RNA-Seq) of these PF Th17 cells revealed significantly up-regulated genes and down-regulated genes in stage I-II and stage III-IV endometriosis, compared with their counterparts in normal PF. In conclusion, this study provides a novel method to isolate live Th17 cells from endometriosis patients, unveils an array of differentially expressed genes in endometriosis Th17 cells, and offers valuable gene expression profile information for endometriosis clinical research.


Subject(s)
Ascitic Fluid/immunology , Endometriosis/immunology , Th17 Cells/physiology , Adult , Female , Gene Expression Regulation , Humans , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, CXCR3/genetics , Receptors, Chemokine/genetics , Sequence Analysis, RNA
14.
J Reprod Immunol ; 149: 103462, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34915278

ABSTRACT

Endometriosis (EDT), a common estrogen-dependent inflammatory disorder, is characterized by endometrial-like tissue outside the uterus. While its pathogenesis is poorly understood, it is supposed that the immune system plays a role in its pathophysiology, and increased number of immune cells and changes in both cell-mediated and humoral immunity have been described. Dendritic cells (DCs) are antigen-presenting cells (APC) of the immune system that recognize, capture, and process complex antigens and present them to T cells, conferring them a unique ability as mediators between the innate and adaptive immune systems. This systematic review aims to enlighten possible disturbances (systemically and locally) of DCs in the development and progression of endometriosis. A search using the strategy: ("dendritic cells" AND "immunology" AND "endometriosis") in databases resulted in 490 citations; after applying inclusion and exclusion criteria, a total of 13 studies were assessed. The evaluated studies demonstrated that DCs are susceptible to pro-endometriotic changes which could inhibit immature DCs (imDCs) from their maturation and induce imDCs into a macrophage phenotype. In addition, the growth and vascularization of endometriosis requires the presence of endogenous DC, which infiltrate endometriotic lesions and enhance endothelial cell migration by secreting proangiogenic factors. Whereas DC maturation suppresses this response, imDC actively promote angiogenesis and growth, leading to a switch in their immunologic role from presenting antigens to support angiogenesis and EDT progression.


Subject(s)
Dendritic Cells/immunology , Endometriosis/immunology , Endothelial Cells/physiology , Animals , Antigen Presentation , Cell Differentiation , Female , Humans , Neovascularization, Pathologic
15.
Am J Reprod Immunol ; 87(1): e13502, 2022 01.
Article in English | MEDLINE | ID: mdl-34592011

ABSTRACT

PROBLEM: Innate lymphoid cells (ILCs), a recently discovered family of innate immune cells, are responsible for the early immune response, and control both innate and adapted immune system via cytokine secretion. The role of ILCs in endometriosis has not been investigated; therefore, here, we aimed to investigate how the proportion of ILCs changes in endometriosis. METHOD OF STUDY: The percentage of each ILC group in CD45+ cells was examined in the peripheral blood, peritoneal fluid, endometrium, and ovarian endometrioma obtained from women with and without endometriosis (ERB-C-1216) using flow cytometry. RESULTS: Specimens were obtained from 19 women with endometriosis and 15 without endometriosis. In the endometrium, patients with endometriosis had lower proportion of ILC2 and 3 compared to control specimens (ILC2: .02±.01% vs .07±.03%; P < .05, ILC3: .31±.14% vs 1.10±.93%; P < .05). There was no significant change in the peripheral blood or the peritoneal fluid between the two groups. Additionally, ovarian endometrioma increased the proportion of ILCs (ILC1: .92±1.12%, ILC2: .08±.08%, ILC3: .70±.39%) compared to the endometrium samples of patients with endometriosis each with P < .05. Immunohistochemistry of IL-1ß and IL-23, which are ILC3-inducing factors, showed no significant change in the H-score of the epithelium of the two groups, but a significant increase was found in ovarian endometrioma. CONCLUSION: The proportion of ILC2 and 3 was reduced in the endometrium of patients with endometriosis, and ILCs were increased in ovarian endometrioma. Our findings may indicate a new immunological approach to understand the pathophysiology of endometriosis.


Subject(s)
Endometriosis/immunology , Endometrium/immunology , Immunity, Innate/physiology , Lymphocytes/pathology , Adult , Cytokines/immunology , Endometriosis/pathology , Endometrium/pathology , Female , Humans , Middle Aged , Young Adult
16.
Fertil Steril ; 117(1): 160-168, 2022 01.
Article in English | MEDLINE | ID: mdl-34656305

ABSTRACT

OBJECTIVE: To characterize T lymphocyte infiltration and programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) expression in early-stage endometriosis-associated ovarian cancer (EAOC), ovarian endometriosis (OE), atypical endometriosis (AE), and deep endometriosis (DE). DESIGN: Case-control, retrospective study. SETTING: Research University Hospital. PATIENT(S): A total of 362 patients with a histologic diagnosis of EAOC, OE, AE, or DE were identified between 2000 and 2019 from Fondazione Policlinico Universitario Agostino Gemelli IRCCS and Gemelli Molise SpA tissue data banks. A 1:1 propensity score-matched method yielded matched pairs of 55 subjects with EAOC, 55 patients with OE, 12 patients with AE, and 42 patients with DE, resulting in no differences in family history of cancer, parity, and use of oral contraceptives. INTERVENTION(S): Immunohistochemistry assays using the following primary antibodies: CD3+; CD4+; CD8+; PD-1; and PD-L1. MAIN OUTCOME MEASURE(S): To characterize T lymphocyte infiltration and PD-1/PD-L1 expression in 4 different endometriosis-related diseases. RESULT(S): Endometriosis-associated ovarian cancer cases displayed significantly higher levels of PD-1/PD-L1 expression compared with all other endometriosis-related diseases (vs. OE vs. AE vs. DE). Moreover, a significantly lower count of infiltrating T lymphocytes was observed in EAOC cases compared with OE ones. Finally, one-third of OE cases showed a cancer-like PD-1/PD-L1 expression profile. CONCLUSION(S): Endometriosis-associated ovarian cancer is characterized by higher levels of PD-1/PD-L1 expression compared with benign endometriosis-related diseases. This profile was found in one-third of clinically benign cases, suggesting that it develops early in the carcinogenesis process.


Subject(s)
Endometriosis/complications , Ovarian Neoplasms/etiology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/pathology , Adult , B7-H1 Antigen/metabolism , Carcinoma, Ovarian Epithelial/etiology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/metabolism , Case-Control Studies , Cell Movement/immunology , Chemotaxis, Leukocyte/physiology , Endometriosis/immunology , Endometriosis/metabolism , Female , Humans , Immunohistochemistry , Italy , Middle Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Retrospective Studies , T-Lymphocytes/physiology
17.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34884907

ABSTRACT

Endometriosis (EM) is an estrogen-dependent disease characterized by the presence of epithelial, stromal, and smooth muscle cells outside the uterine cavity. It is a chronic and debilitating condition affecting ~10% of women. EM is characterized by infertility and pain, such as dysmenorrhea, chronic pelvic pain, dyspareunia, dysuria, and dyschezia. Although EM was first described in 1860, its aetiology and pathogenesis remain uncertain. Recent evidence demonstrates that the peripheral nervous system plays an important role in the pathophysiology of this disease. Sensory nerves, which surround and innervate endometriotic lesions, not only drive the chronic and debilitating pain associated with EM but also contribute to a growth phenotype by secreting neurotrophic factors and interacting with surrounding immune cells. Here we review the role that peripheral nerves play in driving and maintaining endometriotic lesions. A better understanding of the role of this system, as well as its interactions with immune cells, will unearth novel disease-relevant pathways and targets, providing new therapeutics and better-tailored treatment options.


Subject(s)
Endometriosis/immunology , Nerve Growth Factors/metabolism , Neurogenic Inflammation/etiology , Endometriosis/complications , Female , Gene Expression Regulation , Humans , Neurogenic Inflammation/immunology , Pelvic Pain/etiology , Pelvic Pain/immunology , Sensory Receptor Cells/immunology
18.
Front Immunol ; 12: 752646, 2021.
Article in English | MEDLINE | ID: mdl-34691070

ABSTRACT

Background: Endometriosis is a common gynecological disease characterized by the presence of endometrial tissue outside the uterus causing chronic inflammation, severe pain, and infertility. However, the innate immunity of gamma-delta (γδ) T lymphocytes in endometriosis has not been characterized. Women with endometriosis present numerous endocrine and immune dysfunctions and elevated risk for endometrial, ovarian, and breast cancers. The tyrosine kinase EphA2 is often overexpressed in cancer including endometrial carcinoma. Methods: We analyzed Vδ1 and Vδ2 γδ T cells in peripheral blood and paired peritoneal fluid samples in endometriosis patients (n = 19) and compared the counts with that of age- and sex-matched healthy donors (n = 33) using flow cytometry. Vδ1 and Vδ2 T cells isolated from healthy donors were used against KLE, RL-95, and Ishikawa endometrial tumor cells in 4 h flow cytometric cytotoxicity assays. The EphA2 blocking studies were performed using antibody, small-molecule inhibitor ALW-II-41-27, and the CRISPR/Cas9. Results: We determined Vδ1 T cells substantially reduced in patients' peripheral blood (p < 0.01) and peritoneal fluid (p < 0.001). No differences were found for circulating Vδ2 T cells compared with peritoneal fluid samples. We observed inherent cytotoxic reactivity of Vδ1 and Vδ2 γδ T lymphocytes against endometrial tumor cells. Importantly, we found reduced specific lysis of EphA2-positive cell lines KLE and RL-95 by Vδ1 T cells in the EphA2 antibody blocking studies and by the EphA2 inhibitor. Furthermore, Vδ1 T-cell-mediated killing was significantly decreased in RL-95 cell EPHA2 knockout. Finally, potent cytolytic activity exerted by Vδ1 T cells was significantly reduced in EPHA2 knockouts in renal A-498 and colon HT-29 carcinoma cell lines. Conclusions: We determined variable levels of Vδ1 and Vδ2 γδ T cells in endometriosis patients. We observed inherent cytotoxic reactivity of γδ T-cell subsets against endometrial cell lines. Specifically, we found that blocking of EphA2 expression resulted in significant inhibition of endometrial tumor killing mediated by Vδ1 γδ T cells. These results suggest that EphA2 is involved in tumor cell lysis and contributes to susceptibility to Vδ1 γδ T cells cytotoxic reactivity.


Subject(s)
Endometrial Neoplasms/immunology , Endometriosis/immunology , Intraepithelial Lymphocytes/immunology , Receptor, EphA2/metabolism , Adult , Cell Line, Tumor , Endometrial Neoplasms/metabolism , Endometriosis/metabolism , Female , Humans , Middle Aged
19.
Front Immunol ; 12: 749979, 2021.
Article in English | MEDLINE | ID: mdl-34630429

ABSTRACT

Endometriosis is an estrogen-dependent gynecological disease. The pathogenesis of endometriosis remains controversial, although it is generally accepted that the inflammatory immune response plays a crucial role in this process. Mast cells (MCs) are multifunctional innate immune cells that accumulate in endometriotic lesions. However, the molecular mechanism by which estrogen modulates MCs in the development of endometriosis is not well understood. Here we report that estrogen can induce the expression of NOD-like receptor family pyrin domain containing 3 (NLRP3) through estrogen receptor (ER)-α via the estrogen responsive element (ERE) in MCs. Such transcriptional regulation is necessary for the activation of NLRP3 inflammasome and the production of mature interleukin (IL)-1ß in MCs. Targeted inhibition of NLRP3 significantly restrained lesion progression and fibrogenesis in a mouse model of endometriosis. Collectively, these findings suggest that MCs contribute to the development of endometriosis through NLRP3 inflammasome activation mediated by nuclear-initiated estrogen signaling pathway.


Subject(s)
Endometriosis/immunology , Inflammasomes/immunology , Mast Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Cell Line , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/genetics , Interleukin-1beta/immunology , Mast Cells/drug effects , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritoneal Cavity/cytology , Signal Transduction/drug effects , Thiazolidines/pharmacology , Thiones/pharmacology
20.
Mol Hum Reprod ; 27(11)2021 11 02.
Article in English | MEDLINE | ID: mdl-34643696

ABSTRACT

Evidence accumulated in recent years has revealed that neutrophils are involved in the initial establishment of endometriosis, which is well-known as a chronic inflammatory disease. So far, why and how neutrophils promote the formation of early endometriosis are still unclear. In this study, using a mouse model of endometriosis, we demonstrated that endometriosis mice (EMs mice) had a significantly increased number of neutrophils in peritoneal fluids and lesions, and increased levels of granulocyte colony-stimulating factor (G-CSF) and IL-6 in serum and peritoneal fluids compared to the control group. In the neutrophils and uterine fragments co-injection experiment, neutrophils regulated by G-CSF and IL-6 had a similar effect to neutrophils from EMs mice, increasing the number, area, weight and microvessel density (MVD) of endometriotic lesions. Blocking the effect of G-CSF and IL-6 in EMs mice resulted in a decrease in the number, area and weight of endometriotic lesions. Following the depletion of neutrophils in vivo using a anti-Ly6G antibody, the MVD in the lesions of mice treated with neutrophils from EMs mice and neutrophils from pG/pI6 mice were significantly reduced. Neutrophils from EMs mice and neutrophils from pG/pI6 mice altered the expression levels of Mmp9, Bv8 and Trail genes compared to the neutrophils from PBS-treated mice. IL-6 together with G-CSF induced a higher expression of phospho-STAT3 and STAT3 in neutrophils. These findings suggest that neutrophils modulated by G-CSF and IL-6 through the STAT3 pathway alter the expression levels of the angiogenesis-related genes Mmp9, Bv8 and Trail, and may promote the establishment of early endometriosis.


Subject(s)
Endometriosis/metabolism , Endometrium/blood supply , Endometrium/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/metabolism , Neovascularization, Pathologic , Neutrophils/metabolism , Animals , Disease Models, Animal , Endometriosis/immunology , Endometriosis/pathology , Endometrium/immunology , Female , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Interleukin-6/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Microvascular Density , Neuropeptides/genetics , Neuropeptides/metabolism , Neutrophils/immunology , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...