Objective: To evaluate effect of age and parity on distribution and number of cells expressing major histocompatibility complex (MHC) class II, CD4, or CD8 molecules in the endometrium of mares during estrus.

Animals: 32 gynecologically healthy mares, categorized as young (3 to 8 years; n = 17) or old (9 to 16 years; 15) and nulliparous (n = 6), nulliparous embryo donors (16), or parous (10).

Procedures: Endometrial specimens collected from the uterine body and horns during estrus were stained by use of the avidin-biotin-peroxidase method, using monoclonal antibodies against equine MHC class II, CD4, and CD8 molecules. Labeled cells in the stratum compactum within 5 randomly selected fields at 400x magnification (total area = 0.31 mm2) were counted, and numbers were compared among groups and between locations.

Results: Age did not affect cell numbers within the 3 cell subsets examined. Numbers in each subset were higher in the uterine body than in the horns, although the difference was not significant for cells expressing MHC class II. Significantly more cells expressing MHC class II molecules were detected in the uterine body of nulliparous and parous mares than in embryo donors, whereas in the horns, these cells were significantly higher in number only in parous mares. Parity did not affect number of CD4+ or CD8+ cells.

Conclusions And Clinical Relevance: The increased likelihood for endometritis to develop in mares as they age cannot be explained by a decrease in number of cells expressing MHC class II, CD4, or CD8 molecules within the endometrium. However, greater number of cells within these 3 subsets detected in the uterine body, compared with the horns, during estrus suggests a local readiness to act against microorganisms or semen introduced during mating or insemination.

Download full-text PDF

Source

Publication Analysis

Top Keywords

cells expressing
20
mhc class
20
class cd4
16
cd4 cd8
16
cd8 molecules
16
uterine body
16
molecules endometrium
12
number cells
12
expressing mhc
12
age parity
8

Similar Publications

Clinical evidence increasingly suggests that traditional treatments for dysfunctional uterine bleeding (DUB) have limited success. In this study, blood samples from 10 DUB patients and 10 healthy controls were collected for transcriptome sequencing. Then, the differentially expressed genes (DEGs) were screened and crossed with the DUB-related module genes to obtain the target genes.

View Article and Find Full Text PDF

Lysyl oxidase (LOX), a copper-containing secretory oxidase, plays a key role in the regulation of extracellular stiffness through cross-linking with collagen and elastin. Among the LOX family of enzymes, LOX-like 4 (LOXL4) exhibits pro-tumor and anti-tumor properties; therefore, the functional role of LOXL4 in tumor progression is still under investigation. Here, we first determined that transforming growth factor-β1 (TGF-β1) significantly decreased LOXL4 expression in human breast cancer MDA-MB-231 cells, which suggested that decreased LOXL4 may participate in tumor progression.

View Article and Find Full Text PDF

Background: Ovarian cancer (OC), particularly high-grade serous ovarian carcinoma (HGSOC), is the leading cause of mortality from gynecological malignancies worldwide. Despite the initial effectiveness of treatment, acquired resistance to poly(ADP-ribose) polymerase inhibitors (PARPis) represents a major challenge for the clinical management of HGSOC, highlighting the necessity for the development of novel therapeutic strategies. This study investigated the role of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a pivotal regulator of glycolysis, in PARPi resistance and explored its potential as a therapeutic target to overcome PARPi resistance.

View Article and Find Full Text PDF

Background: Ovarian cancers (OC) and cervical cancers (CC) have poor survival rates. Tumor-infiltrating lymphocytes (TILs) play a pivotal role in prognosis, but shared immune mechanisms remain elusive.

Methods: We integrated single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) to explore immune regulation in OC and CC, focusing on the PI3K/AKT pathway and FLT3 as key modulators.

View Article and Find Full Text PDF

Background: Pathogenic or null mutations in WRN helicase is a cause of premature aging disease Werner syndrome (WS). WRN is known to protect somatic cells including adult stem cells from premature senescence. Loss of WRN in mesenchymal stem cells (MSCs) not only drives the cells to premature senescence but also significantly impairs the function of the stem cells in tissue repair or regeneration.

View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!