Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Dec 1.
Published in final edited form as: Fertil Steril. 2019 Dec;112(6):1118–1128. doi: 10.1016/j.fertnstert.2019.08.060

MACROPHAGES DISPLAY PRO-INFLAMMATORY PHENOTYPES IN EUTOPIC ENDOMETRIUM OF WOMEN WITH ENDOMETRIOSIS WITH RELEVANCE TO AN INFECTIOUS ETIOLOGY OF THE DISEASE

Júlia Vallvé-Juanico a,b,c, Xavier Santamaria b,c,d, Kim Chi Vo a, Sahar Houshdaran a,, Linda C Giudice a,†,*
PMCID: PMC6944306  NIHMSID: NIHMS1545447  PMID: 31843088

Abstract

Objective:

To phenotype transcriptomically macrophages M1 (Mφ1) and M2 (Mφ2) in the endometrium of women with endometriosis.

Design:

Prospective experimental study.

Setting:

University research laboratory.

Patient(s):

Six women with endometriosis and five controls without disease, in the secretory phase of the menstrual cycle.

Intervention(s):

Mφ1, Mφ2, uterine natural killer (uNK) and regulatory-T (Treg) cells were isolated from human endometrium using a uniquely designed cell-specific fluorescence activating cell sorting panel. Transcriptome profiles were assessed by RNA-high sequencing, bioinformatic and biological pathway analyses.

Main Outcomes Measure(s):

Differential gene expression between Mφ1 and Mφ2 in women with and without endometriosis, and in Mφ1 versus Mφ2 in each group, were determined and involved different bioloigcal and signaling pathways.

Result(s):

Flow cytometry analysis showed no significant differences in total numbers of leukocytes between control and endometriosis groups, although Mφ1 were higher in endometriosis versus controls. Statistical transcriptomic analysis was performed only in Mφ1 and Mφ2 populations due to larger sample sizes. Bioinformatic analyses revealed that in women with endometriosis endometrial Mφ1 are more pro-inflammatory than controls and Mφ2 paradoxically have a pro-inflammatory phenotype.

Conclusion(s):

As Mφ are phenotypically plastic and their polarization state depends on their microenvironment, the altered endometrial environment in women with endometriosis may promote endometrial Mφ2 polarization and an Mφ1 pro-inflammatory phenotype. Moreover, aberrant phenotypes of Mφ may contribute to abnormal gene expression of eutopic endometrium and a pro-inflammatory environment in women with endometriosis relevant to the pathophysiology of the disease and compromised reproductive outcomes.

Keywords: endometriosis, macrophages, inflammation, endometrium, infection

CAPSULE

RNA-Seq analyses showed that macrophages-M1 are more pro-inflammatory in eutopic endometrium of women with endometriosis than in control endometrium and that macrophages-M2 paradoxically display a pro-inflammatory phenotype in disease.

INTRODUCTION

Endometriosis is an estrogen-dependent inflammatory disease that results in pelvic pain and/or infertility. It affects approximately 10% of reproductive age women (14) and is characterized by the presence of endometrial-like tissue outside the uterus where it elicits an inflammatory response (1,3,4). The eutopic endometrium of women with endometriosis has been widely studied with regard to dysfunctionality of steroid hormone response, stem cell populations, and recruitment of immune populations for immune tolerance and overall tissue homeostasis and pregnancy success versus women without disease (13). However, there are scant data about the function and phenotypes of eutopic endometrial immune cells in women with and without endometriosis. As the endometrial immune niche involves multiple cell types with varying degrees of activation and communications among immune and non-immune cells that dictate functionality of the tissue, characterizing the endometrial immune niche is of great relevance to understanding endometrial function and dysfunction.

Uterine natural killer cells (uNK) secrete angiogenic factors that contribute to the maturation of blood vessels having a role in embryo implantation and successful pregnancy (6,7). In healthy endometrium, their cytotoxic activity diminishes during the secretory phase of the menstrual cycle which allows embryo implantation (810). However, in infertile endometriosis patients, uNK have high cytotoxicity in eutopic endometrium that could lead to an inhospitable environment for embryo implantation (11). Other immune cells, such as T regulatory cells (Treg), have been also described to behave differently in the endometrium of women with endometriosis. In healthy endometrium, they increase in the proliferative and decrease during the secretory phase, with the latter creating an immune-tolerant environment allowing embryo implantation. However, in infertile women with endometriosis, Treg are increased in the peri-implantation endometrium, leading to an implantation failure (12).

Endometriosis has been referred to as “a disease of the macrophage” (13), based mainly on a replete literature on the roles and functionality of this cell type in peritoneal fluid of women with disease and in establishment of endometriosis lesions and associated processes of angiogenesis and fibrosis. Mφ are key effector cells in both innate and humoral immunity as they phagocytose pathogens, act as antigen presenting cells, and have a role in tissue regeneration, angiogenesis and wound healing (14). In eutopic endometrium of women without endometriosis, their numbers vary throughout the menstrual cycle, increasing in the secretory and menstrual phases (15). This increase may be attributed to their phagocytic properties and role in clearing cell debris and apoptotic cells during endometrial shedding (16). Cycle variation among endometrial Mφ does not occur in women with endometriosis (17), suggesting that survival of shed and refluxed endometrial cells may be enhanced, enabling them to migrate to the peritoneal cavity and establish disease. Mφ are classified as either “classically activated” Mφ (Mφ1) or “alternatively activated” Mφ (Mφ2) (14) and, depending on the microenvironment, they can switch from one type to the other (18). Mφ1 have a role in pro-inflammatory responses; whereas, Mφ2 are involved in angiogenesis, anti-inflammatory reactions, and tissue repair (14,19). In healthy endometrium, the predominant population is Mφ2 (19,20), suggesting that the normal environment is anti-inflammatory. Taken together, most of the studies in eutopic endometrium have focused on the number of immune cells in this tissue and how they fluctuate throughout the cycle, but little is known about their functionality in women with endometriosis.

Herein, we designed a novel flow cytometry panel to isolate Mφ1, Mφ2, Treg and uNK from eutopic endometrium of women with endometriosis and those with no evidence of disease. RNA High-Sequencing (RNA-Seq) was used to elucidate Mφ1 and Mφ2 phenotypes and possible functions in disease. Overall, the data support a phenotypic switch of the common anti-inflammatory Mφ2 to the pro-inflammatory Mφ1 phenotype and a more exaggerated pro-inflammatory phenotype of the Mφ1 population in women with endometriosis.

MATERIALS AND METHODS

Sample Collection and Processing

Eleven endometrial biopsies in the secretory phase were collected: 6 from women with endometriosis (stage I-IV) and 5 from women with no evidence of endometriosis at the time of the surgery for benign gynecologic disorders. The mean age was 37 and 42 (23-49) years old, respectively. In order to evaluate if the age could confound the results, a non-parametric t-test with a subsequent Mann-Whitney test (p<0.05) was performed and no significant differences between groups were found (p=0.2641). Patients had not used hormonal therapy for at least three months prior to the study. Endometrial samples were obtained through the University of California San Francisco (UCSF) NIH Human Endometrial Tissue and DNA Bank under approval of the UCSF Committee on Human Research (IRB#10-02786), and written informed consent was obtained from all participants. Endometrial tissue was digested as previously described (21). Briefly, it was minced mechanically and incubated for one hour at 37ºC in digestion media, which contained collagenase type I and hyaluronidase (21). Subsequently, the single cell suspension was filtered using a 40μm mesh to discard cell clumps, and single cells were cryopreserved in liquid nitrogen until use.

Flow cytometry panel design

A cytometry panel of 10 conjugated antibodies able to separate the immune populations of interest (Mφ1, Mφ2, Treg and uNK) was designed. Specific membrane markers of resident and blood infiltrating immune cells were included to avoid sorting cells derived from the peripheral circulation. The brightest colors were used for the markers with the lowest antigen density. Minimum overlapping of 11 colors (10 antibodies plus the live/dead dye) was achieved. The markers and lasers used for each population are in Table 1 and the gating strategy is in Figure 1.A. First, the cells were gated with CD45 (leukocyte marker), conjugated with brilliant violet 605 (BV605). In the case of Mφ, usually this population is a resident tissue population, thus no specific tissue markers were used. It is challenging to differentiate between the Mφ1 and Mφ2 subpopulations, as they have some common markers and have the ability to polarize from one type to the other. Our strategy was as follows: for both types, CD14 marker conjugated with phycoerythrin (PE) was used. For Mφ2, CD163, a specific marker for this population, was used conjugated with PE-cyanin7 (PE-Cy7). As there are no specific markers for Mφ1 and it was suspected that the concentration of activated Mφ1 would be low in the endometrium (as Mφ2 are higher than Mφ1 in normal endometrium (19)), CD80 (activation marker) conjugated with peridinin chlorophyll protein complex-cyanin 5.5. (PerCP-Cy5.5) antibody, a bright dye, was used. With regard to Treg, they express CD3 and CD4 markers. The most accepted specific marker for Treg is Foxp3. This, is an intracellular marker and thus could not be used for sorting. However, they also express CD25. To be able to discern between tissue Treg and Treg deriving from circulation, CD69, an activation marker for Treg that is expressed in tissue, was included. Thus, CD3+CD4+CD25+CD69+ cells (tissue Treg) were isolated. The CD3 antibody was conjugated with ultraviolet B 737 (BUV737), as was the CD4 antibody with ultraviolet B 395 (BUV395). CD25 has a low antigen density, therefore, brilliant blue 515 (BB515) was used, which is one of the brightest dyes. Then, CD69, a tissue activation Treg marker, was conjugated with allophycocyanin-cyanin7 (APC-Cy7), assuring that only resident Treg were isolated. For uNK cells, which are CD56+, as it is known that blood NK are CD16+; whereas, uNK are CD16Low/− (2225), CD56+CD16 were collected. For CD56, brilliant violet 421 (BV421), was used and in the case of CD16, allophycocyanin (APC) conjugated antibody was used. Finally, to separate between live and dead cells, Aqua dye was used (sources of all antibodies are listed in Table 1).

Table 1. Antibodies used for the FACS.

The table shows all the markers used for the sorting of the desired populations, the fluorochrome to which they were conjugated, the antibody, the distributor, and the laser with which the fluorochromes are excited. Excitation and emission wavelenghts for each fluorechrome are also shown.

Marker Fluorochrome (excitation/emission) Conjugated Antibody Manufacturer* and reference FACS Aria Laser

Live/dead Aqua (405nm/512nm) - ThermoFisher (L34957) Violet E
CD45 BV605 (405nm/600nm) CD45-BV605 BD Bioscience (BDB 564047) Violet
CD14 PE (496nm/578) CD14-PE Biolegend (301805) YGD
CD163 PE-Cy7 (561nm/785nm) CD163-PE-Cy7 Biolegend (333614) YGA
CD80 PerCP-Cy5.5 (488nm/695nm) CD80-PerCP-Cy5.5 Biolegend (305231) Blue A
CD3 BUV737 (355nm/737nm) CD3-BUV737 BD Bioscience (BDB 564307) UVA
CD4 BUV395 (355nm/395nm) CD4-BUV395 BD Bioscience (BDB 563550) UVC
CD25 BB515 (488nm/515nm) CD25-BB515 BD Bioscience (564468) Blue B
CD69 APC-Cy7 (640nm/785nm) CD69-APC-Cy7 BD Bioscience (BDB 560737) RedA
CD56 BV421 (405nm/421nm) CD56-BV421 BD Bioscience (562752) Violet F
CD16 APC-Cy7 (640nm/660nm) CD16-APC Biolegend (302011) Red C
*

ThermoFisher, Waltham, MA, USA; BD BioScience, East Rutherford, NJ, USA; Biolegend, San Diego, CA, USA.

Figure 1. Cytometry panel and flow cytometry analyses.

Figure 1.

A. Gating strategy. The different gates used to sort the four desired populations are shown. The X and Y axis show the lasers that were used to gate the cells. In addition, the name of the marker and the fluorochrome used have been inserted on the corresponding axes. First, cells were gated by their complexity (SSC-A/FSC-A). Then, singles cells were gated (FSC-W/FSC-H) as well as live cells (L-Aqua Violet E/FSC-A). In this case, two different gates were made: one for T cells and uNK and a bigger one for Mφ due to the autofluorescence present in the latter. The next step was to gate live CD45+ cells (L-Aqua Violet E/CD45 BV605 Violet D). Thereafter, three strategies were used; 1) gating resident Treg (CD45+CD3+CD4+CD25+CD69+); 2) gating uNK (CD45+CD3CD56+CD16), and 3) gating Mφ2 (CD3CD14+CD163+) and Mφ1 (CD3CD14+CD163CD80+). The final sorted populations are marked with a yellow star. B. Number of sorted cells. The table shows the number of cells of macrophages M1 (Mφ1), macrophages M2 (Mφ2), resident regulatory T cells (Treg) and uterine natural killer (uNK) cells, obtained after FACS in non-endometriosis and endometriosis patients. C. Percentage of CD45+ cells. The figure shows that no significant differences were found in the % CD45+ cells between control (n=5) and endometriosis patients (n=6). D. Percentage of each subpopulation comparions between control and endometriosis. Mφ1 increased significantly in numbers in endometriosis endometrium; whereas, no significant differences were found for Mφ2, uNK and Treg. E. Percentage of uNK and Treg coming from blood circulation. It can be observed that in both control (left panel) and endometriosis (right panel) endometrium, there is a significant increase of resident uNK (CD16) compared to circulating NK (CD16+), which indicates that there is almost no NK contamination from the peripheral circulation. In the case of Treg, it can be observed that there is an increase of Treg coming from blood in both groups, altough it it is not significant.

Fluorescence Activated Cell Sorting (FACS)

Endometrial samples were thawed at 37ºC. After centrifugation at 1,300rpm for five minutes, the supernatant was discarded and the pellet was washed with 1X PBS. After another centrifugation, cells were resuspended with 1X PBS + 5% bovine serum albumin (BSA) and incubated at room temperature for 30 minutes. A minimum of 100,000 unlabeled cells were separated as a negative control. Ten conjugated antibodies were used to label the samples (Table 1). 1μl of antibody per million cells was used in all cases except for CD45 and CD4, where 2μl per million cells were needed for an optimal cell labelling. A solution of all Fluorochromes Minus One (FMO) was prepared for each antibody to assess any overlap among the channels in the FACS instrument. After one hour of incubation at 4ºC in 1X PBS + 3% BSA and in the dark, cells were washed with 1X PBS and centrifuged for 5 minutes at 1,300 rpm. The pellet was resuspended with 500μl of 1X PBS and labelled with 1μl of LIVE/DEAD™ Fixable Aqua Dead cell labelling dye (ThermoFisher, Waltham, MA). On the other hand, UltraComp eBeads compensation magnetic beads (ThermoFisher, Waltham, MA, USA) were labelled with each of the 10 antibodies following the manufacturer’s instructions, to allow the correction of the spectral overlap between fluorochromes. Using the gating strategy (Figure 1.A), each population was sorted in the FACS Aria Jabba the Hutt (BD Biosciences, East Rutherford, NJ, USA) instrument and collected in 1X PBS. Flow cytometry analysis of the sorted cells was performed using FlowJo.v10 software (FlowJo LLC, Ashland, OR, USA), and statistical analyses (Mann-Whitney test, p-value<0.05) were conducted using GraphPad software (GraphPad Software Inc, San Diego, CA, USA).

RNA extraction

As low yields of cells were obtained after FACS (Figure 1.B), RNeasy micro kit (Qiagen, Hilden, Germany) was used to isolate RNA from Mφ populations (note yields from uNK and Treg cells were too low, and were not used for RNA-Seq, see Methods). From the 22-sorted Mφ samples (Mφ1 and Mφ2 populations of each of the 11 endometrial samples), RNA was extracted following the manufacturer’s instructions to perform the total RNA-seq library prep from samples containing >900 cells (9 samples). The library preparation from the remaining samples, which contained at least 20 cells, was performed directly from cells in 1X PBS. RNA was eluted in 10μl of RNase free-water, and the quality (RNA integrity numbers (RIN)) and concentration were measured using a Tapestation4200 System (Agilent, Santa Clara, CA, USA).

RNA-Seq and statistical and bioinformatic analyses

SMART-Seq™ v4 Ultra™ low input RNA kit for sequencing (Clontech, Mountain View, CA, USA) was used to perform the RNA-seq library preparation. It allows RNA-Seq to be performed with very low concentrations of RNA or to use whole cells to preserve sample integrity. In total, library preparations for 22 samples (10 from control and 12 from endometriosis) were performed. The quality of fastq files was tested using the FastQC (v0.11.5) (26) and the Qualimap (rnaseq module – v2.2.1) software (27). Reads were aligned with the STAR mapper (v2.5.2a) (28) to release 88 of the Homo sapiens ENSEMBL version of the genome (GRCh38/hg38 assembly) (29). A raw count of reads per gene was also obtained with STAR (28) . In order to overcome the heterogeneity between samples, first, samples were removed from the analysis if they had <5 million uniquely mapped reads, and the remaining samples were downsampled to 30 million mapped reads when needed. The data have been deposited in NCBI GEO database (accession number GSE130435). The R/Bioconductor package DESeq2 (v1.20.0) (3032) was used to assess differential expression between experimental groups (Wald statistical test + false discovery rate (FDR) correction). Statistically significant differentially expressed genes (DEG) were considered when FDR<0.05 and log fold change>2 (LogFC>2). Different comparisons performed using Mφ populations are shown in Table 2. Biological significance analyses were conducted using Ingenuity Pathway Analyses (IPA) software (Ingenuity® Systems, Redwood City, CA, USA), and significant molecular functions were established with an activation Z-score> |2.00|.

Table 2. Comparisons of the Mφ populations analyzed by RNA-seq.

The first column shows each comparison performed after quality control analyses. The second column shows the number of samples used in each population to perform each comparison. The last column shows the number of differentially expressed genes obtained in each comparison by Wald statistical test and FDR. Mφ1: macrophages 1; Mφ2: macrophages 2; DEG: differentially expressed genes; LogFC: log fold change; FDR: false discovery rate.

Comparisons Sample size DEG (LogFC≥2/FDR<0.05)

1. Mφ Endo vs. Mφ Control 11 vs. 7 1567
2. Mφ1 Control vs. Mφ2 Control 3 vs. 4 1260
3. Mφ1 Endo vs. Mφ2 Endo 5 vs. 6 705
4. Mφ1 Endo vs. Mφ1 Control 5 vs. 3 1422
5. Mφ2 Endo vs. Mφ2 Control 6 vs. 4 1544

RESULTS

FACS and flow cytometry analyses

After FACS, low cell numbers were obtained (Figure 1.B) that subsequently guided further analyses. Cytometry analyses from all the immune populations showed that CD45+ cells corresponded to an average of 6.8% of the total sample, in agreement with other studies wherein leukocytes comprise 10-20% of total endometrial cells (3338). No significant differences in CD45+ cells were observed between control and endometriosis groups (Figure 1.C). Statistical analyses comparing percentage of each sub-population between controls and endometriosis were performed, and no significant differences were observed except for Mφ1, which was significantly higher in the endometriosis group (p=0.0087) (Figure 1.D). Because resident tissue markers were included in the cytometry panel, contamination of immune populations from the peripheral circulation could also be calculated. In both the control and endometriosis groups, uNK (CD16) were significantly higher compared to blood NK (CD16+) (Figure 1.E), demonstrating that there was almost no contamination with blood NK cells. The percentage of Treg coming from blood (CD69) was higher than tissue Treg (CD69+), although it was not significant (Figure 1.E). Due to the low number of uNK and Treg cells obtained, RNA-Seq was only performed in the Mφ populations, that had significantly greater numbers of cells. Thus, from the 44 original FACS-sorted immune populations, 22 samples (Mφ populations) from endometriosis and control were used for the transcriptome study.

RNA extraction and gene expression analyses (RNA-Seq)

RNA concentrations extracted from Mφ ranged between 5-45ng/μl. After RNA-Seq and quality controls, we excluded any FastQ sequences for which the number of reads did not reach our threshold of five million reads/sequence. Thus, the populations analyzed were: 5 Mφ1 endometriosis, 3 Mφ1 control, 6 Mφ2 endometriosis and 4 Mφ2 control. After statistical analysis, DEG (FDR<0.05 and LogFC≥2) were found in all comparisons (Table 2; Supplemental Table 1).

Biological significance of the DEG analyses revealed significant molecular functions, relevant molecules secreted by Mφ1 and Mφ2, activation/inhibition of upstream regulators and de-regulated networks in each comparison (activation Z-score≥2.00) (Table 3). The 25 top de-regulated networks are in Supplemental Table 2. Increase in cell-cell contact was observed along with repression of RNA molecular functions, when comparing Mφ1 endometriosis versus Mφ1 control (Comparison 4, Table 2). Increased cell-cell contact is consistent with, e.g., increased adhesion to bacteria to accomplish bacterial engulfment. Top de-regulated networks showed overexpression of cellular development, growth and proliferation, and overexpression of immune response-related networks, such as infectious disease and antimicrobial and inflammatory responses (Table 3). These data indicate that Mφ1 in endometriosis have a more extensive pro-inflammatory phenotype than Mφ1 in the control group.

Table 3. De-regulated molecular functions, networks and upstream regulators in macrophages comparisons.

The table shows the significant molecular de-regulated molecular functions (Z-Score≥2) obtained after IPA analyses of different macrophages comparisons, as well as the diferentially expressed genes of RNA-Seq dataset involved in these functions. The fifth column shows the five top de-regulated networks in each comparison, where the immune-related networks are bolded. Finally, the sixth column shows the upstream regulators predicted to be activated (↑) or inhibited (↓) in the IPA analysis of each comparison. Mφ1: Macrophages 1; Mφ2: Macrophages 2; Endo: Endometriosis; Ctr: Control; ERG: ETS-related gene; TNF: Tumor Necrosis Factor; LH: Lutein hormone; miR-483-3p: micoRNA-483-3p; TGFB1: Transforming Growth Factor Beta 1; SATB: Stabilin 1; INHBA: Inhibin Subunit Beta A; false discovery rate. NFkB: Nuclear Factor kappa Beta; IFNα: Interferon alpha; IL15: Interleukin 15; VCAN: Versican.

Molecular Functions Z-Score Genes De-regulated Networks Upstream regulators Z-Score
Mφ-Endo vs Mφ Ctr Senescence of fibroblast cell line

Activation of cells
−2.09

1.95
ARHGAP10, DPY30, EREG, HYAL1, IL1A, LATS1, MAPK1, MATN4, ME2, PBRM1, POT1, PTEN, PTTG1, RBL1

ANGPT1, C3, CADM1, CD1D, CD4, CHRNA7, CSF2, DDIT4, EGF, EIF3A, GP1BA, HLAB1, IL13, IL1A, IL1R1, IL33, ITGA3, LBP, LTA, MYO18A, NOS3, NT5E, PSEN1, PTEN, PTGDR2, PTPN22, RAB5B, SBNO2, SIRPG, SPI1, STIM1, THBS1, TOP2A, VAMP4, VEGFA, WASL
Hematological System Development and Function, Immune Cell Trafficking, Inflammatory Response

Infectious Diseases, Antimicrobial Response, Inflammatory Response

Organismal Injury and Abnormalities, Cell Morphology, Cellular Development

Cellular Development, Cellular Growth and Proliferation, Reproductive System Development and Function

Dermatological Diseases and Conditions, Inflammatory Disease, Inflammatory Response
↑ERG 2,333
Molecular Functions Z-Score Genes Z-Score
Mφ1 Endo vs Mφ1 Ctr
Cell-cell contact

Repression of RNA
2.25

2.19
ACTN4, ANGPT1, ARF6, ARHGAP19, CBLL1, CLDN2, CTNNB1, DNM2, DSP, ESRP2, F2RL2, GDF15, GSK3B, IL13, ING4, ITGA4, NRDC, NT5E, PLCB1, RAPGEF3, SYK, TJP1

CUL3, DR1, ERCC2, FOXG1, LCOR, MECP2, TAF1
Cellular Development, Cellular Growth and Proliferation, Lymphoid Tissue Structure and Development

Infectious Diseases, Antimicrobial Response, Inflammatory Response

Cardiac Arrythmia, Cardiovascular Disease, Hereditary Disorder

Cell Morphology, Cellular Movement, Organismal Injury and Abnormalities

Cell Signaling, Cellular Assembly and Organization, Cellular Function and Maintenance
↑TNF 2,587
Molecular Functions Z-Score Genes Z-Score
Mφ2 Endo vs Mφ2 Ctr Concentration of Ca2+

Transport of carbohydrates

Internalization of bacteria
2.36

2.21

2.15
DRD1, ITGAL, KLRD1, LPAR4, PLG, TRPV5, VIPR2

ABCB1, AQP2, C3, CSF2RA, NPC1, RALBP1, SLC23A1, SLC2A3, SLC45A1, SLC5A1

C3, CEACAM6, ERBB2, PLG, PRKCA
Connective Tissue Disorders, Inflammatory Disease, Inflammatory Response

Drug Metabolism, Endocrine System Development and Function, Lipid Metabolism

Cell-To-Cell Signaling and Interaction, Hematological System Development and Function, Immune Cell Trafficking

Cell Death and Survival, Hematological System Development and Function, Lymphoid Tissue Structure and Development

Behavior, Nervous System Development and Function, Endocrine System Disorders
↓LH −2,169
Molecular Functions Z-Score Genes Z-Score
Mφ1 vs Mφ2 Ctr
Cytotoxicity of leukocytes
−2.18 BMPR1A, CBLB, CD244, FCAR, KIR2DL4, KIR3DL1, KLRC1, NOTCH2 Cancer, Cell Death and Survival, Hematological Disease

Cellular Function and Maintenance, Cellular Movement, Carbohydrate Metabolism

Cell Death and Survival, Cancer, Organismal Injury and Abnormalities

Developmental Disorder, Hereditary Disorder, Organismal Injury and Abnormalities

Hematological System Development and Function, Lymphoid Tissue Structure and Development, Tissue Morphology
↓miR-483-3p

↑TGFB1
−2,000

2,195
Molecular Functions Z-Score Genes Z-Score
Mφ1 vs Mφ2 Endo Growth of S. Cerevisiae

Engulfment of cells
−2.00

2.12
LIG1, PARP1, POLR2K, ZMPSTE24

AP2B1, ATP6V1E1, AXL, CD47, CDC5L, CERK, CSK, DNM2, EGR1, ERBB2, LMBRD1, NR1H3, RHOG, SNX5, STK4, SYK, USPL1
Cell-To-Cell Signaling and Interaction, Hematological System Development and Function, Immune Cell Trafficking

Cell Death and Survival, Embryonic Development, Organismal Injury and Abnormalities

Cell Death and Survival, Inflammatory Response, Cancer

Cellular Assembly and Organization, Cellular Function and Maintenance, Neurological Disease

Cancer, Cell Death and Survival, Organismal Injury and Abnormalities
↑TNF

↓SATB1

↑INHBA

↑NFkB

↑IFNα

↑IL15

↓VCAN
2,385

−2,400

2,449

2,784

2,186

2,224

−2,000

In contrast, molecular functions upregulated in Mφ2 in endometriosis (Comparison 5, Table 2) included an accumulation of Ca2+, increase in carbohydrate transport, and internalization of bacteria (Table 3). When comparing Mφ1 of women with versus without endometriosis (comparison 4, Table 2), the upstream regulator TNFα was predicted to be increased (Table 3). Increased internalization of bacteria is consistent with phagocytic properties of the pro-inflammatory Mφ1 phenotype. The top networks in endometrial Mφ2 from women with endometriosis included deregulation of connective tissue disorders, endocrine system development and function, lipid metabolism, inflammatory disease/response, and drug metabolism (Table 3). These data overall demonstrate that Mφ2 in eutopic endometrium of women with endometriosis have a pro-inflammatory phenotype, compared to Mφ2 in control women.

DISCUSSION

In the current study, we developed a cytometry panel that allowed for separating circulating immune cells and tissue resident cells and different immune cell types within human endometrium. Thus, the analyzed immune populations were purely tissue-activated resident cells devoid of contamination by circulating immune cells. One goal was to develop and optimize this panel for the current study. However, it will also have value for other researchers aiming to separate these tissue-specific populations, since it is a challenging panel to design due to the multiple colors used and the possible overlap between channels.

After cytometry analyses, where Mφ1 were found to be significantly higher in endometriosis, Mφ were studied in more detail by transcriptomic analyses. To our knowledge this is the first RNA-Seq dataset of Mφ in eutopic endometrium of women with endometriosis. Abnormal distribution of Mφ within eutopic endometrium of women with disease could contribute to the aberrant distribution of immune cells in the pelvic cavity and the abnormal development and gene expression of this tissue. While Mφ maintain organ homeostasis and facilitate host defense and wound healing, they also underlie the pathogenesis of many chronic inflammatory diseases (39).

The increased de-regulated molecular functions and networks in Mφ1 in endometrium of women with endometriosis indicate these cells have a more pro-inflammatory phenotype than Mφ1 in the control group. In addition, a significantly higher number of sorted Mφ1 was observed in endometriosis patients (Figure 1.D), confirming a previous report (40). Moreover, these results suggesting that eutopic endometrium of women with endometriosis is more pro-inflammatory than control endometrium, is consistent with findings from other groups (17,41).

An unexpected finding herein was the pro-inflammatory phenotype exhibited by endometrial Mφ2 from women with endometriosis. Mφ2 in other tissues generally display an anti-inflammatory phenotype (39), and, importantly, Mφ are phenotypically plastic with regard to their polarization state depending on their microenvironment (42). Moreover, Mφ1 and Mφ2 gene expression signatures often overlap, and the resultant phenotype depends on the tissue microenvironment (40). Thus, endometrial Mφ2 of women with endometriosis could undergo polarization in situ to Mφ1, adopting a pro-inflammatory phenotype, due to an altered environment. The paradigm of different subpopulations of Mφ is controversial in the immunology literature. Specifically, it is unclear whether there are unique Mφ populations (as Mφ1, Mφ2) or if Mφ comprise a unique population that alters its phenotype depending on environmental cues. Herein, we have referred to Mφ as two different subpopulations (Mφ1 and Mφ2), although the dynamics and mechanisms driving pro-inflammatory and anti-inflammatory Mφ functional phenotypes remain to be determined.

Notably, tumors take advantage of macrophage plasticity. For example, in the early phases of cancer, high production of Mφ1 inflammatory mediators activates the adaptive immune response capable of eliminating nascent neoplastic cells, and also support neoplastic transformation (40). In contrast, once the tumor is stablished, the main population of Mφ is Mφ2, producing an anti-inflammatory environment, which allows tumor growth. Endometriosis it is not a malignancy, however, it shares some characteristics with cancers. In endometriotic lesions and peritoneal fluid of women with endometriosis, e.g., Mφ2 are increased (43), indicating that, as in cancer, an anti-inflammatory environment prevails favoring development and growth of the endometriotic lesions. In addition, that Mφ2 have a role in angiogenesis further supports this paradigm. Finally, Mφ2 are also involved in nerve growth, suggesting they may also have a role in endometriosis-related pain (44).

The initial pro-inflammatory phenotype of Mφ in cancer increases NFKB and downstream events and increases transcription of pro-inflammatory cytokines such as TNFα, IL12, IL23, IL1β, IL6, and ROS. In the current study, the NFKB pathway was activated in Mφ1 of endometriosis, which does not occur in Mφ1 of control women (Table 3). Indeed, it has been described that the NFKB pathway is de-regulated in the eutopic endometrium of women with endometriosis (45), which also indicates that the microenvironment in endometrium of women with disease is more pro-inflammatory than heathy tissue.

Notably, an increase of transport of carbohydrates was observed in Mφ2 of women with endometriosis. It is known that glycolysis is high in Mφ1 and is decreased in Mφ2 and Mφ polarization may derive from a reprogramming of glucose metabolism (46). Several studies have suggested that altering nutrient availability or blocking specific metabolic pathways skews the Mφ phenotype and alters their effector functions in chronic inflammatory diseases (47). In this regard, Mφ metabolism modulation could open a new therapeutic window for treating inflammatory diseases including endometriosis.

Finally, the upstream regulator TNFα was increased in IPA analysis when comparing Mφ1 of women with versus without endometriosis (comparison 4, Table 2), as well as increased Ca2+ accumulation was activated in Mφ2 (Table 3). It has been noted that a transient increase of Ca2+ plays a role in the expression of TNFα by Mφ1 (48). Intracellular Ca2+ oscillations are likely to induce permanent changes in Mφ physiology, and a supra-physiologic elevation of Ca2+ in mitochondria can be cytotoxic and induce apoptosis in the long term (48). Whether TNFα-mediated events play a role in Mφ function awaits further studies.

Over the past decade, high-throughput sequencing techniques have challenged the dogma of the sterility of the uterine endometrium (4954), and in particular an altered endometrial microbiome in women with endometriosis has been proposed (55). In addition, the endometrial microbiome also correlates with IVF outcomes (52), although whether this occurs in women with endometriosis awaits further study. However, treatment with antibiotics resulted in reduced numbers of endometriosis lesions in a mouse model, with concomitant alteration of the gut microbiome (56), although the endometrial microbiome was not reported in this study (54). Interestingly, a recent systematic review supports the use of antibiotics prior to oocyte retrieval in patients with endometriosis, among other gynecologic disorders (57). The presence of pathogenic, non-commensal bacteria in the endometrium may induce an altered immune cell profile and activation (increased numbers and activation of Mφ1 and activation of Mφ2) that could impact the production of cytokines by immune resident cells that adversely affect embryo implantation (58). In addition to effects on reproductive outcomes, the observed greater pro-inflammatory endometrial environment herein could be related to the pathophysiology of the disease. While attractive, we are aware that the sample size of the study is small. Therefore, these results should be taken with caution. Finally, whether the pro-inflammatory phenotype of the Mφ2 population reported herein is in response to commensal bacteria or pathogens, or if Mφ populations are implicated in reproductive outcomes, is not clear. However, it is anticipated that this important area of research could have profound implications clinically and diagnostically.

CONCLUSIONS

Overall, the results of the current study lead to the conclusion that both Mφ1 and Mφ2 in eutopic endometrium of women with endometriosis display a higher pro-inflammatory phenotype compared to controls without disease. Endometrial Mφ2 appear to be predisposed to Mφ1 polarization in women with endometriosis, thus increasing their inflammatory phenotype. These findings suggest that eutopic endometrium has different Mφ gene signatures depending on the presence or absence of disease and that the endometrial environment of women with endometriosis is more pro-inflammatory than control endometrium. Whether subtypes of the disease are associated with different subsets of immune and whether the macrophage pro-inflammatory status is related to bacteria in the endometrium of women with disease are yet to be determined. Finally, the results herein may have implications regarding the impact of the macrophage phenotypes on reproductive outcomes and possible novel therapeutics for microbiome-related symptoms and response for fertility and pain in women with endometriosis.

Supplementary Material

1
2

ACKNOWLEDGMENTS

Funding was provided by NIH NICHD HD055764-12 (LCG), the Kerfuffle Foundation (LCG), SRI International Training Grant (JVJ), Industrial Doctorate ID-2015-074 (AGAUR, Spain) (JVJ), NIH S10 1S10OD021822-01 supporting the Aria Fusion “Jabba the Hutt” in UCSF Cytometry Core, and JVJ acknowledges Sarah Bonnin from Centre for Genomic Regulation (Barcelona) for her help in the bioinformatics analyses.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

BIBLIOGRAPHY

  • 1.Giudice LC. Clinical Practice: Endometriosis. N Engl J Med 2010;362:2389–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Becker CM, Laufer MR, Stratton P, Hummelshoj L, Missmer SA, Zondervan KT, et al. World Endometriosis Research Foundation Endometriosis Phenome and Biobanking Harmonisation Project: I. Surgical phenotype data collection in endometriosis research. Fertil Steril 2014;102:1213–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.K. JJ, L BA. Endometrial Receptivity in Eutopic Endometrium of Women with Endometriosis It is affected, let me show you why. Fertil Steril 2017;108:19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zondervan KT, Becker CM, Koga K, Missmer SA, Taylor RN, Viganò P. Endometriosis. Nat Rev Dis Prim 2018;4:9. [DOI] [PubMed] [Google Scholar]
  • 5.Burney RO, Giudice LC. Pathogenesis and pathophysiology of endometriosis. Fertil. Steril 2012;98:511–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li XF, Charnock-jones DS, Zhang EKO, Hiby S, Malik S, Day K, et al. Angiogenic Growth Factor Messenger Ribonucleic Acids in Uterine Natural Killer Cells. J Clin Endocrinol Metab 2015;86:1823–34. [DOI] [PubMed] [Google Scholar]
  • 7.Lysakova-Devine T, O’Farrelly C. Tissue-specific NK cell populations and their origin. J Leukoc Biol 2014;96:981–90. [DOI] [PubMed] [Google Scholar]
  • 8.Salamonsen LA, Lathbury LJ. Endometrial leukocytes and menstruation. Hum Reprod Update 2000;6:16–27. [DOI] [PubMed] [Google Scholar]
  • 9.Yovel G, Shakhar K, Ben-Eliyahu S. The Effects of Sex, Menstrual Cycle, and Oral Contraceptives on the Number and Activity of Natural Killer Cells. Gynecol Oncol 2001;81:254–62. [DOI] [PubMed] [Google Scholar]
  • 10.Souza SS, Castro FA, Mendonça HC, Palma PVB, Morais FR, Ferriani RA, et al. Influence of menstrual cycle on NK activity. J Reprod Immunol 2001;50:151–9. [DOI] [PubMed] [Google Scholar]
  • 11.Giuliani E, Parkin KL, Lessey BA, Young SL, Fazleabas AT. Characterization of uterine NK cells in women with infertility or recurrent pregnancy loss and associated endometriosis. Am J Reprod Immunol 2014;72:262–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen S, Zhang J, Huang C, Lu W, Liang Y, Wan X. Expression of the T regulatory cell transcription factor FoxP3 in peri-implantation phase endometrium in infertile women with endometriosis. Reprod Biol Endocrinol 2012;10:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Capobianco A, Rovere-Querini P. Endometriosis, a disease of the macrophage. Front Immunol 2013;4:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677–86. [DOI] [PubMed] [Google Scholar]
  • 15.Salamonsen LA, Zhang J, Brasted M. Leukocyte networks and human endometrial remodelling. J Reprod Immunol 2002;57:95–108. [DOI] [PubMed] [Google Scholar]
  • 16.Khan KN, Masuzaki H, Fujishita A, Kitajima M, Sekine I, Matsuyama T, et al. Estrogen and progesterone receptor expression im macrophages and regulation of hepatocyte growth factor by ovarian steroids in women with endometriosis. Hum Reprod 2005;20:2004–13. [DOI] [PubMed] [Google Scholar]
  • 17.Berbic M, Schulke L, Markham R, Tokushige N, Russell P, Fraser IS. Macrophage expression in endometrium of women with and without endometriosis. Hum Reprod 2009;24:325–32. [DOI] [PubMed] [Google Scholar]
  • 18.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005;5:953–64. [DOI] [PubMed] [Google Scholar]
  • 19.Jensen AL, Collins J, Shipman EP, Wira CR, Guyre PM, Pioli PA. A Subset of Human Uterine Endometrial Macrophages is Alternatively Activated. Am J Reprod Immunol 2012;68:374–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cominelli A, Gaide Chevronnay HP, Lemoine P, Courtoy PJ, Marbaix E, Henriet P. Matrix metalloproteinase-27 is expressed in CD163+/CD206+M2 macrophages in the cycling human endometrium and in superficial endometriotic lesions. Mol Hum Reprod 2014;20:767–75. [DOI] [PubMed] [Google Scholar]
  • 21.Barragan F, Irwin JC, Balayan S, Erikson DW, Chen JC, Houshdaran S, et al. Human Endometrial Fibroblasts Derived from Mesenchymal Progenitors Inherit Progesterone Resistance and Acquire an Inflammatory Phenotype in the Endometrial Niche in Endometriosis. Biol Reprod 2016;94:118–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.King A Uterine leukocytes and decidualization. Hum. Reprod. Update. 2000;6:28–36. [DOI] [PubMed] [Google Scholar]
  • 23.Koopman LA, Kopcow HD, Rybalov B, Boyson JE, Orange JS, Schatz F, et al. Human Decidual Natural Killer Cells Are a Unique NK Cell Subset with Immunomodulatory Potential. J Exp Med 2003;198:1201–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Salamonsen LA, Woolley DE. Menstruation: Induction by matrix metalloproteinases and inflammatory cells. J. Reprod. Immunol 1999;44:1–27. [DOI] [PubMed] [Google Scholar]
  • 25.Yang Z, Kong B, Mosser DM, Zhang X. TLRs, macrophages, and NK cells: Our understandings of their functions in uterus and ovary. Int Immunopharmacol 2011;11:1442–50. [DOI] [PubMed] [Google Scholar]
  • 26.Andrews S FastQC - A quality control tool for high throughput sequence data. 2010.
  • 27.Okonechnikov K, Conesa A, Garcia-Alcalde F. Qualimap 2: Advanced multi-sample quality control for high-throughput sequencing data. Bioinformatics 2015;32:292–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ftp://ftp.ensembl.org/pub/release-88/fasta/homo_sapiens. ENSEMBL: Homo sapiens, release 88.
  • 30.Huber Wolfgang et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat Methods 2015;12:115–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Foundation for Statistical Computing, Vienna A. R Core Team (2014). R: A language and environment for statistical computing. http://www.R-project.org/.
  • 32.Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Evans J, Salamonsen LA. Inflammation, leukocytes and menstruation. Rev Endocr Metab Disord 2012;13:277–88. [DOI] [PubMed] [Google Scholar]
  • 34.Flynn L, Byrne B, Carton J, O’Farrelly C, Kelehan P, O’Herlihy C. Menstrual Cycle Dependent Fluctuations in NK and T-Lymphocyte Subsets from Non-Pregnant Human Endometrium. Am J Reprod Immunol 2000;43:209–17. [DOI] [PubMed] [Google Scholar]
  • 35.Givan AL, White HD, Stern JE, Colby E, Gosselin EJ, Guyre PM, et al. Flow cytometric analysis of leukocytes in the human female reproductive tract: comparison of fallopian tube, uterus, cervix, and vagina. Am J Reprod Immunol 1997;38:350–9. [DOI] [PubMed] [Google Scholar]
  • 36.King AE, Critchley HOD. Oestrogen and progesterone regulation of inflammatory processes in the human endometrium. J Steroid Biochem Mol Biol 2010;120:116–26. [DOI] [PubMed] [Google Scholar]
  • 37.Wira CR, Rodriguez-Garcia M, Patel MV. The role of sex hormones in immune protection of the female reproductive tract. Nat Rev Immunol 2015;15:217–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yeaman G, Collins J. CD8 + T cells in human uterine endometrial lymphoid aggregates : evidence for accumulation of cells by traficking. Immunology 2001;2:434–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Parisi L, Gini E, Baci D, Tremolati M, Fanuli M, Bassani B, et al. Review Article Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J Immunol Res 2018;2018: 8917804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Takebayashi A, Kimura F, Kishi Y, Ishida M, Takahashi A, Yamanaka A, et al. Subpopulations of Macrophages within Eutopic Endometrium of Endometriosis Patients. Am J Reprod Immunol 2015;73:221–31. [DOI] [PubMed] [Google Scholar]
  • 41.Vallvé-Juanico J, Suárez-Salvador E, Castellví J, Ballesteros A, Hugh ST et al. Aberrant expression of epithelial leucine-rich repeat containing G protein-coupled receptor 5–positive cells in the eutopic endometrium in endometriosis and implications in deep-infiltrating endometriosis. Fertil Steril 2017;108:858–867. [DOI] [PubMed] [Google Scholar]
  • 42.Das A, Sinha M, Datta S, Abas M, Chaffee S, Sen CK, et al. Monocyte and Macrophage Plasticity in Tissue Repair and Regeneration. Am. J. Pathol 2015;185:2596–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, et al. Macrophages Are Alternatively Activated in Patients with Endometriosis and Required for Growth and Vascularization of Lesions in a Mouse Model of Disease. Am J Pathol 2009;175:547–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Morotti M, Vincent K, Brawn J, Zondervan KT, Christian M. Peripheral changes in endometriosis-associated pain. Hum Reprod Updat 2015;20:717–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhou B, Rao L, Peng Y, Wang Y, Qie M, Zhang Z, et al. A Functional Promoter Polymorphism in NFKB1 Increases Susceptibility to Endometriosis. DNA Cell Biol 2010;29:235–9. [DOI] [PubMed] [Google Scholar]
  • 46.Blagih J, Jones RG. Polarizing macrophages through reprogramming of glucose metabolism. Cell Metab 2012;15:793–5. [DOI] [PubMed] [Google Scholar]
  • 47.Di Conza G, Ho PC. Metabolic adaptation of macrophages in chronic diseases. Cancer Lett 2018;414:250–6. [DOI] [PubMed] [Google Scholar]
  • 48.Clapham DE. Calcium signaling. Cell 1995;80:259–68. [DOI] [PubMed] [Google Scholar]
  • 49.Kyono K, Hashimoto T, Kikuchi S, Nagai Y, Sakuraba Y. A pilot study and case reports on endometrial microbiota and pregnancy outcome: An analysis using 16S rRNA gene sequencing among IVF patients, and trial therapeutic intervention for dysbiotic endometrium. Reprod Med Biol 2019;18:72–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Verstraelen H, Vilchez-Vargas R, Desimpel F, Jauregui R, Vankeirsbilck N, Weyers S, et al. Characterisation of the human uterine microbiome in non-pregnant women through deep sequencing of the V1-2 region of the 16S rRNA gene. PeerJ 2016;4:e1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fang R-L, Chen L-X, Shu W-S, Yao S-Z, Wang S-W, Chen Y-Q. Barcoded sequencing reveals diverse intrauterine microbiomes in patients suffering with endometrial polyps. Am J Transl Res 2016;8:1581–92. [PMC free article] [PubMed] [Google Scholar]
  • 52.Moreno I, Codoner FM, Vilella F, Valbuena D, Martinez-Blanch JF, Jimenez-Almazan J, et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am J Obstet Gynecol 2016;215:684–703. [DOI] [PubMed] [Google Scholar]
  • 53.Baker JM, Chase DM, Herbst-Kralovetz MM. Uterine microbiota: Residents, tourists, or invaders? Front Immunol 2018;9:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Franasiak JM, Scott RT. Endometrial microbiome. Curr Opin Obstet Gynecol 2017;29(3):146–52. [DOI] [PubMed] [Google Scholar]
  • 55.Khan KN, Fujishita A, Hiraki K, Kitajima M, Nakashima M, Fushiki S, et al. Bacterial contamination hypothesis: a new concept in endometriosis. Reprod Med Biol 2018;17:125–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Chadchan SB, Cheng M, Parnell LA, Yin Y, Schriefer A, Mysorekar IU, et al. Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: a potential role for gut microbiota. Hum Reprod 2019;1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pereira N, Hutchinson AP, Lekovich JP, Hobeika E, Elias RT. Antibiotic Prophylaxis for Gynecologic Procedures prior to and during the Utilization of Assisted Reproductive Technologies: A Systematic Review. J Pathog 2016;2016:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sirota I, Zarek SM, Segars JH. Potential influence of the microbiome on infertility and assisted reproductive technology. Semin Reprod Med 2014;32:35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES