Described herein is a protocol to isolate and analyze the infiltrating leukocytes of tissues at the maternal-fetal interface (uterus, decidua, and placenta) of mice. This protocol maintains the integrity of most cell surface markers and yields enough viable cells for downstream applications including flow cytometry analysis.
Immune tolerance in pregnancy requires that the immune system of the mother undergoes distinctive changes in order to accept and nurture the developing fetus. This tolerance is initiated during coitus, established during fecundation and implantation, and maintained throughout pregnancy. Active cellular and molecular mediators of maternal-fetal tolerance are enriched at the site of contact between fetal and maternal tissues, known as the maternal-fetal interface, which includes the placenta and the uterine and decidual tissues. This interface is comprised of stromal cells and infiltrating leukocytes, and their abundance and phenotypic characteristics change over the course of pregnancy. Infiltrating leukocytes at the maternal-fetal interface include neutrophils, macrophages, dendritic cells, mast cells, T cells, B cells, NK cells, and NKT cells that together create the local micro-environment that sustains pregnancy. An imbalance among these cells or any inappropriate alteration in their phenotypes is considered a mechanism of disease in pregnancy. Therefore, the study of leukocytes that infiltrate the maternal-fetal interface is essential in order to elucidate the immune mechanisms that lead to pregnancy-related complications. Described herein is a protocol that uses a combination of gentle mechanical dissociation followed by a robust enzymatic disaggregation with a proteolytic and collagenolytic enzymatic cocktail to isolate the infiltrating leukocytes from the murine tissues at the maternal-fetal interface. This protocol allows for the isolation of high numbers of viable leukocytes (>70%) with sufficiently conserved antigenic and functional properties. Isolated leukocytes can then be analyzed by several techniques, including immunophenotyping, cell sorting, imaging, immunoblotting, mRNA expression, cell culture, and in vitro functional assays such as mixed leukocyte reactions, proliferation, or cytotoxicity assays.
Immune tolerance in pregnancy is a period when distinctive changes occur within the immune system of the mother. These changes allow the mother to tolerate the fetus, a semi-allogenic graft1. The fetus expresses paternal major histocompatibility complex (MHC) antigens2, and fetal cells have been found in the maternal circulation3; however, the fetus is not rejected4,5. This enigma is not fully understood.
The most recent hypothesis states that maternal-fetal tolerance is created during coitus and fecundation6,7 and maintained to sustain a full-term pregnancy8-10. A breakdown of this maternal-fetal tolerance is considered a mechanism of disease during early and late stages of pregnancy10-16. Maternal-fetal tolerance involves the participation of various leukocyte sub-populations, including T cells (regulatory T cells, Th1 cells, Th2 cells, and Th17 cells), macrophages, neutrophils, mast cells, NK cells, and NKT cells, dendritic cells, and B cells, that change in density and localization throughout pregnancy15,17-19. Maternal-fetal tolerance is enriched at the maternal-fetal interface20 – the anatomical site where the immune system of the mother interacts with the fetal antigens20,21.
The maternal-fetal interface is created during placentation when the fetal extravillous trophoblast cells invade the uterine mucosa22-24. On the fetal side of this interface, the membranes surrounding the fetus create a specialized epithelial surface within the placenta, and the syncytiotrophoblast cells control the nutrient exchange through their direct contact with maternal blood22. On the maternal side of the interface, the decidua recruits a heterogeneous pool of leukocytes that in mice account for 30% to 50% of all decidual cells. In addition to their participation in maternal immune tolerance, these cells are key contributors to different processes during pregnancy, e.g., the protection of the reproductive tract from infections, fecundation, embryo implantation7,25, decidual angiogenesis26, vascular remodeling24,27, trophoblast invasion28, placental development24,25, and, ultimately, labor and delivery15,17. Therefore, the study of the leukocytes involved in maternal-fetal tolerance is essential to elucidating the pathogenesis of pregnancy-related complications.
While the use of immunohistochemistry and immunofluorescence has generated data for the direct visualization and localization of uterine, decidual, or placental leukocytes29,30, flow cytometry analysis has further revealed specific subsets of leukocytes in each of these tissues31,32. Additionally, flow cytometry has been used to determine the density and proportion of maternal-fetal interface leukocytes33 and expression levels of extracellular and intracellular proteins8-10,34. Flow cytometric analysis of leukocytes at the maternal-fetal interface requires a single-cell suspension. In order to isolate infiltrating leukocytes from the decidual, uterine, and placental tissues, two methods of tissue dissociation have been used: mechanical and enzymatic. Both methods allow the separation of infiltrated leukocytes from the extracellular matrix (ECM) of these tissues. Enzymatic tissue dissociation is superior to mechanical tissue dissociation as it allows a higher yield of leukocytes with less shear-force-associated damage35. Consequently, mechanical tissue dissociation requires pooling tissues36, which may increase the variability and heterogeneity of the samples. Yet, mechanical dissociation may be the choice when the antigen of interest can be altered by enzymatic dissociation or when the functionality of the cells of interest need to be preserved (e.g., cytotoxicity of NK cells)35.
The use of proteolysis with specific enzymes to degrade the ECM eliminates the low yields observed with mechanical dissociation. Several studies have reported the use of trypsin32, collagenase37, DNase31, dispase38, and commercial cocktails of various enzymes32,39. However, the nature and concentration of different enzymes and the duration of digestion must be meticulously defined and validated in order to ensure maintenance of the integrity of the cell surface antigenic epitopes required for immunophenotyping. The various surface structures are differentially susceptible to destruction by different enzymes, with some enzymes, such as trypsin, being notorious for stripping leukocyte surface epitopes recognized by many monoclonal antibodies.
Introduced herein is a method using a proteolytic and collagenolytic enzymatic cocktail, called Accutase. This enzymatic solution is gentle enough while still efficient in dissociating murine tissues at the maternal-fetal interface, and does not require the addition of other dissociating reagents or serum to terminate the dissociation reaction. Moreover, it is ready to use as supplied and, although the time of dissociation needs to be validated, it is more robust than the above-noted enzymes40,41.
The utilization of a combination of both types of tissue disaggregation improves the quality and amount of cells obtained; thus, several studies have implemented the combined use of mechanical and enzymatic dissociation with satisfactory results31,32,37. The protocol described herein was established and validated in our laboratory; it uses a combination of a gentle mechanical dissociation followed by a robust enzymatic disaggregation. This protocol allows the isolation and further study of the infiltrating leukocytes in murine tissues at the maternal-fetal interface (uterus, decidua, and placenta). The following protocol also maintains the integrity of cell surface markers and yields enough viable cells for downstream applications as demonstrated by flow cytometric analysis. Finally, this protocol maintains the consistency of cell preparation for the analysis and comparison of different murine tissues composing the maternal-fetal interface.
Before working with the samples mentioned in this protocol, animal ethical approval must be given by the Local Research Ethics Committee and the Institutional Review Boards. When working with animal blood, cells, or hazardous agents as mentioned in this protocol, the proper biosafety and laboratory safety actions must be followed.
1. Mouse Handling and Tissue Collection
2. Uterine, Decidual, and Placental Tissue Dissociation
3. Viability Staining for Fixable Cells
4. Immunophenotyping
5. Viability Staining for Unfixable Cells
6. Cell Culture
7. Magnetic Cell Sorting
The dissection of murine tissues from the maternal-fetal interface is shown in Figure 1; this procedure includes opening the peritoneal cavity (Figure 1A,B), uterine horns (Figure 1C) including the implantation sites (Figure 1D), and the collection of the uterine tissues (Figure 1E), placenta (Figure 1F), and decidual tissues (Figure 1G) at 16.5 dpc. Figure 2 shows the morphology of isolated macrophages (F4/80+) collected from the decidual and uterine tissues at 16.5 dpc using magnetic cell sorting. Isolated macrophages maintain the ability to release cytokines (data not shown). The yield of viable cells isolated from the decidua, uterus, and placenta is shown in Figure 3, and cell viability is greater than 70% in all tissues. Figure 4 shows the gating strategy for analyzing polymorphonuclear and mononuclear leukocytes within the singlets and viability gates, including T cells (CD45+CD3+), neutrophils (CD45+Ly6G+), macrophages (CD45+F4/80+), dendritic cells (CD45+CD11c+), and NK cells (CD45+CD49b+) at 16.5 dpc. A high proportion of macrophages co-express CD11c. Figure 5 shows neutrophils, macrophages, and T cells in the decidual, uterine, and placental tissues at 16.5 dpc. Figure 6 shows the gating strategy for analyzing lymphocytes within the singlets and viability gates, including T cells (CD45+CD3+) and B cells (CD45+B220+) at 16.5 dpc. T cells include CD4+ and CD8+ T cells. Murine tissues at the maternal-fetal interface also include CD3+CD4-CD8- (gamma-delta T cells) in high proportions. Figure 7 shows T cells and B cells in the decidual, uterine, and placental tissues at 16.5 dpc.
Figure 1. Tissue dissection. (A) Uterine horns at 16.5 dpc in a B6 mouse. The uterine horns are attached to the mesentery and draining vessels. (B) Uterine horns dissected from the mesentery and still attached to the cervix. (C) Uterine horns including the implantation sites and cervix. (D) Dissection of an implantation site. (E) Dissection of the uterine tissues from the implantation site. (F) Separation of the placenta and decidua from the implantation site. (G) Detachment of the decidua (white-gray layer) from the placenta. Please click here to view a larger version of this figure.
Figure 2. Macrophages isolated from decidual and uterine tissues. Macrophages (F4/80+ cells) isolated from the decidual and uterine tissues at 16.5 dpc using magnetic cell sorting. 20X magnification. Please click here to view a larger version of this figure.
Figure 3. Viability of isolated cells. Viable cells (DAPI- cells represented with arrows) isolated from decidual, uterine, and placental tissues. Please click here to view a larger version of this figure.
Figure 4. Gating strategy for polymorphonuclear and mononuclear leukocytes. Total leukocyte population was gated within the singlets and viability gates. T cells (CD45+CD3+), macrophages (CD45+F4/80+), neutrophils (CD45+Ly6G+), dendritic cells (CD45+CD11c+), and NK cells (CD45+CD49b+) were gated within the total-leukocyte gate (CD45+). Please click here to view a larger version of this figure.
Figure 5. Neutrophils, macrophages, and T cells in murine tissues at the maternal-fetal interface. Neutrophils (CD45+Ly6G+), macrophages (CD45+F4/80+), and T cells (CD45+CD3+) were gated within the viability and total-leukocyte (CD45+) gates in isolated decidual, uterine, and placental cells. Please click here to view a larger version of this figure.
Figure 6. Gating strategy for lymphocytes. Mononuclear cells were gated within the singlets and viability gates. T cells (CD3+) and B cells (B220+) were gated within the viability gate. CD4+ and CD8+ T cells were gated within the T-cell gate (CD3+). Please click here to view a larger version of this figure.
Figure 7. Lymphocyte sub-populations in murine tissues at the maternal-fetal interface. T cells (CD3+) and B cells (B220+) were gated within the viability gate in isolated decidual, uterine, and placental cells. CD4+ and CD8+ T cells were gated within the T-cell gate (CD3+). Please click here to view a larger version of this figure.
Cell marker | Fluorochrome | Clone | Company | Catalog Number |
LIVE/DEAD | DAPI | – | Life Technologies | L23105 |
CD45 | V450 | 30-F11 | BD Biosciences | 560501 |
CD3 | FITC | 145-2C11 | BD Biosciences | 553062 |
CD4 | APC | RM4-5 | BD Biosciences | 553051 |
CD8 | PE-CF594 | 53-6.7 | BD Biosciences | 562283 |
B220 | APC-Cy7 | RA3-6B2 | BD Biosciences | 552094 |
F4/80 | PE | BM8 | eBiosciences | 12-4801-82 |
Ly6G | APC-Cy7 | 1A8 | BD Biosciences | 560600 |
CD49b | APC | DX5 | BD Biosciences | 560628 |
CD11c | PE-Cy7 | HL3 | BD Biosciences | 558079 |
Table 1. List of antibodies utilized for leukocyte subset immunophenotyping
The collection of consistent data that records the abundance and phenotypic characteristics of infiltrating leukocytes at the maternal-fetal interface is essential to understanding the pathogenesis of pregnancy-related complications. Several techniques have been described that facilitate the isolation of infiltrating leukocytes from the murine tissues at the maternal-fetal interface throughout pregnancy31,38,39,43-46. However, each technique is different, uses different enzymes or enzyme combinations, requires different dissociation times, does not specify quantities of tissue, and, most importantly, does not always specify the viability of the isolated cells. The protocol described herein allows the isolation of infiltrating leukocytes from the murine tissues at the maternal-fetal interface with high viability, and provides detailed information about the commercial reagents, buffer preparation, tissue quantities, and incubation times validated in the laboratory.
One of the most critical steps of the leukocyte isolation process is the tissue dissociation; this step involves mechanical homogenization and/or enzymatic reactions that can alter the integrity of extracellular proteins used in phenotypic characterization47. The protocol described herein offers a novel approach that combines the use of gentle mechanical and enzymatic tissue dissociation techniques to preserve the integrity of extracellular markers in the leukocytes isolated from the placenta and the decidual and uterine tissues of mice.
Single enzymes and combinations of different enzymes have been used to isolate infiltrating leukocytes from the murine tissues at the maternal-fetal interface31,32,39,44. In many cases, these enzymes that are prepared to specific concentrations by hand in the laboratory may be subject to human error. Here, instead, a ready-to-use purified collagenase/neutral protease cocktail, Accutase, has been implemented in the laboratory; as a commercially available enzyme preparation, it has been shown to provide reliable results in cell culture48. This enzymatic solution is known to effectively detach macrophages from the culture plates without scraping and, most importantly, without losing surface antigens48. This prepared enzyme has also been used to process the digestion of human and animal nervous system tissues, resulting in viable isolated cells that remain sustainable for long periods, which allows their subsequent culture47. Moreover, this enzymatic solution preserves CD24 antigenicity in isolated cells from central nervous system tissues47. When compared to Liberase-1, another cocktail of collagenase and neutral protease, neither Accutase nor Liberase-1 generate free DNA aggregates; however, Accutase is gentler than Liberase-1 during tissue dissociation47. The collagenase/neutral protease cocktail used in this protocol has also demonstrated superiority to trypsin in the preservation of CD44, a cancer stem cell surface marker49. This laboratory’s studies have consistently noted that this enzymatic solution preserves mouse leukocyte surface antigens. Indeed, informative differences have been found in the expression of extracellular markers in macrophages (CD11b+F4/80+ cells), neutrophils (CD11b+Ly6G+ cells), NKT cells (CD3+CD49b+ cells), T cells (CD3+ cells) and B cells (B220+CD19+ cells), including CD4, CD8, CD69, CD25, CD40L, PD1, CD44, CD62L, and CTLA4, and in cytokine release. Therefore, the method described herein is optimal for immunophenotyping of infiltrating leukocytes at the maternal-fetal interface in mice, as shown in the representative results.
One important advantage of this method is the simultaneous determination of several extracellular and intracellular antigens within the viability gate. The dye most used to determine cell viability is propidium iodide (PI); however, its use is limited since it can be used only in combination with FITC. This is because PI cannot be adequately distinguished from most other fluorochromes excited by blue and red lasers50. A solution is to use DAPI50 or any other dye that is excited by UV lasers but not by blue and red lasers commonly used for immunophenotyping. This protocol allows immunophenotyping of viable cells as it includes the use of DAPI50 for unfixable cells or the use of viability dye for fixable cells.
A second important advantage of the protocol described herein is that it allows the isolation of leukocytes with a high yield of viable cells. The representative data shows that 70% to 89% of the isolated cells are viable. This is of great importance as this protocol has allowed the study of the functional properties of the isolated cells from murine tissues at the maternal-fetal interface. For example, cultures of the decidual and uterine macrophages and the study of cytokine release under stimulation have been performed using this method.
To achieve successful results, the application of the described protocol is important when considering the following factors: 1) tissue collection must be performed within 5 to 10 min after opening the peritoneal cavity, and these tissues must be placed on ice to preserve the viability of the isolated cells; 2) mechanical tissue homogenization must be performed using fine-tip scissors and cannot exceed 2 min since longer periods have been demonstrated to reduce the yield of viable cells; 3) the duration of incubation with the enzymatic solution must be less than 1 hr since its activity diminishes after this period of time; 4) the temperature of incubation with the enzymatic solution must be maintained at 37 °C to obtain the optimal activity of this cocktail of enzymes; 5) cell pellet manipulation must be done gently with micropipettes because use of the vortex can damage the integrity of the cells (isolated cells are differentiated and abrupt manipulation can easily reduce their viability); 6) buffer and centrifuge temperatures must be kept at the same temperature as the cell suspension; 7) isolated cells must be processed for immunophenotyping or used immediately as their viability reduces rapidly; and 8) when performing immunophenotyping, samples must be acquired using a flow cytometer immediately for best results.
A limitation of this protocol is the cost of the enzymatic solution, which is more expensive than other enzymes with similar functions, e.g., trypsin, dispase II, and collagenase. However, the advantages that this enzymatic solution displays over and above the described enzymes are superior.
Besides immunophenotyping and cell culture and sorting, the future applications of this protocol are numerous and varied. For example, it is now possible to determine DNA methylation, expression of target genes, in vitro leukocyte functionality (e.g., phagocytosis, cytotoxicity, T-cell proliferation and plasticity assays, etc.), and production of reactive oxygen species in the leukocytes isolated from murine tissues at the maternal fetal interface. Indeed, description of new and rare leukocytes in murine tissues at the maternal-fetal interface can be also performed.
The authors have nothing to disclose.
NGL was supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health. We gratefully acknowledge Maureen McGerty and Amy E. Furcron (Wayne State University) for their critical reading of the manuscript.
Magentic Cell Separation | |||
MS Columns | |||
Cell Separator | |||
30μm pre separation filters | |||
Multistand | |||
15mL safe lock conical tubes | |||
MACS Buffer | (0.5% bovine serum albumin, 2mM EDTA and 1X PBS) | ||
Reagents | |||
Anti-mouse CD16/CD32 | |||
Anti-mouse extracellular antibodies | (Table 1) | ||
Sodium azide | |||
Bovine serum albumin | (BSA) | ||
LIVE/DEAD viability dye | |||
Fixation buffer solution | |||
FACS Buffer | (1% bovine serum albumin, 0.5% sodium azide, and 1X PBS ph 7.2) | ||
Trypan Blue Solution 0.4% | |||
Fetal bovine serum | |||
Additional Instruments | |||
Incubator with shaker | |||
Flow cytometer | |||
Centrifuge | |||
Vacuum system | |||
Incubator | |||
Water bath | |||
Cell counter | |||
Microscope |