Next Article in Journal
Longitudinal Influences on Maternal–Infant Bonding at 18 Months Postpartum: The Predictive Role of Perinatal and Postpartum Depression and Childbirth Trauma
Previous Article in Journal
Real-World Implementation of PRISMA-7 and Clinical Frailty Scale for Frailty Identification and Integrated Care Activation: A Cross-Sectional Study in Northern Italian Primary Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Porcine Peripheral Blood Mononuclear Cells (PBMCs): Methods of Isolation, Cryopreservation, and Translational Applications in Human Studies

by
Magdalena Pietrzak
1,*,
Monika Chaszczewska-Markowska
2 and
Magdalena Zemelka-Wiacek
1
1
Department of Clinical Immunology, Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
2
Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(10), 3432; https://doi.org/10.3390/jcm14103432
Submission received: 7 April 2025 / Revised: 7 May 2025 / Accepted: 11 May 2025 / Published: 14 May 2025
(This article belongs to the Section Clinical Laboratory Medicine)

Abstract

:
Porcine peripheral blood mononuclear cells (pPBMCs) are increasingly recognized as a valuable model in biomedical and translational research, particularly in contexts directly related to human health and disease. Their immunological features, such as the presence of CD4+CD8+ double-positive T cells and cytokine expression patterns, exhibit a notable degree of similarity to human immune cells, making them an attractive tool for studying human-relevant immune responses. This review outlines current methodologies for isolating and cryopreserving pPBMCs, with a focus on maintaining high cell viability and functionality. Key technical considerations, including the optimal use of gradient media, appropriate anticoagulants, and standardized freezing/thawing protocols, are discussed in detail. Furthermore, the article highlights the applications of pPBMCs in various research contexts, including vaccine development, inflammation studies, infection models, and xenotransplantation. A comparative perspective is provided to identify similarities and differences between porcine and human PBMCs, supporting the validity of swine as a translational model. Evidence from pPBMC-based studies has shown predictive value for human outcomes, reinforcing their role as a surrogate system for preclinical investigations. Given their anatomical, physiological, and immunogenetic similarities to humans, porcine PBMCs represent a valuable bridge between basic science and clinical application, playing an increasingly important role in translational medicine.

1. Introduction

Peripheral blood mononuclear cells (PBMC) are widely used as a representative immune cell population for studying immunological responses and mechanisms in health and disease. They serve as a valuable model for analyzing the body’s response to various challenges, such as pathogens or pharmacological interventions. PBMCs represent a heterogeneous population of peripheral blood cells, primarily including lymphocytes (T helper (Th) CD4+, T cytotoxic (Tc) CD8+, gamma–delta (γδ) T cells, T regulatory (Treg), B, natural killer (NK) cells, monocytes, and dendritic cells (DC) [1,2,3,4,5]. Their main functions involve pathogen recognition, the initiation of immune responses, and the regulation of both innate and adaptive immunity.
Among the various species used in immunological research, the pig has emerged as a particularly valuable model organism. The significance of porcine peripheral blood mononuclear cells (pPBMCs) stems from the unique anatomical, genetic, and immunological similarities between pigs and humans. Swine share comparable immune system components and functional responses, making them more translationally relevant than traditional rodent models in many contexts. This is particularly advantageous in the study of complex human diseases, including inflammatory disorders, infectious diseases, and vaccine responses. Furthermore, the relatively large size of pigs and the ease of repeated blood sampling facilitate longitudinal studies and in-depth immunological analyses. These attributes, combined with the practical benefits of cost-effective husbandry and well-established breeding protocols, make swine and their immune cells highly attractive for biomedical and translational research [6,7]. With the growing interest in this model, there is an increasing need for a thorough understanding of the processes related to the isolation, storage, and application of these cells, paving the way for new discoveries in immunology and translational medicine [8,9].
The use of pPBMCs enables the investigation of species-specific immune processes while offering results that often mirror human immunological responses more accurately than other animal models. This has led to their growing application in vaccine development, xenotransplantation research, and the study of zoonotic infections that pose risks to both swine and human populations. Additionally, pPBMCs provide an essential tool for evaluating the efficacy and safety of novel immunomodulatory therapies before moving to human trials. As such, they represent a critical bridge between basic research and clinical application, supporting the development of next-generation treatments and diagnostic tools [10,11].
The isolation of PBMCs from porcine blood is a process that requires precision to obtain a viable cell population for further analyses. Standard approaches rely on density-based separation techniques, allowing for the extraction of lymphocytes and monocytes from other blood components. This method enables the acquisition of a pure fraction of PBMC, which can then be used in in vitro experiments, such as lymphocyte proliferation assays, surface marker expression analysis, or cytokine production evaluation. A key aspect is the adaptation of protocols to the specific physiology of swine, distinguishing these methods from similar procedures used in other species [12].
Equally important is cryopreservation, a process that allows the long-term storage of cells without compromising their viability or immune responsiveness [13]. The selection of appropriate cryoprotective media plays a crucial role in maintaining cell stability under extreme low-temperature conditions.
The application of pPBMCs in scientific research is extensive and includes studies on innate and adaptive immunity mechanisms, testing new therapeutic strategies, and developing biotechnological products such as vaccines and immunomodulators [14,15,16,17]. Their role in modeling immune responses to viral and bacterial infections, as well as in analyzing reactions to environmental stressors, makes them invaluable across multiple fields. Moreover, pPBMCs are utilized in research on chronic diseases and inflammatory conditions, providing insights into processes with potential implications for human health [15,18,19]. Their availability and suitability for experimental manipulation further enhance their appeal as a research model.
This review aims to compile and systematize current knowledge on pPBMCs, with a particular focus on isolation methods, cryopreservation strategies, and their diverse scientific applications. By discussing these aspects, we aim to highlight both the achievements and gaps in existing approaches, thereby pointing to directions for future research.

2. Isolation of pPBMCs

Currently, a standardized protocol for the isolation of pPBMCs has not yet been developed. As a result, methods adapted from human procedures or based on previous comparative studies on porcine cells are commonly used. One of the main challenges during PBMC isolation is sample contamination with granulocytes.
Due to the higher density of granulocytes and erythrocytes compared to mononuclear cells, density gradient centrifugation is the most widely used method for separation. A critical step in this approach involves adding an anticoagulant, such as ethylenediaminetetraacetic acid (EDTA), heparin, or sodium citrate, to the blood sample, followed by dilution with an equal volume of phosphate-buffered saline (PBS), which facilitates the effective isolation of PBMCs [20]. Figure 1 provides a comprehensive overview of the PBMC isolation process.
Various gradient media are available on the market, including Ficoll-Paque™ (Cytiva, GE Healthcare, Marlborough, MA, USA) (density of 1.077 g/mL), Histopaque™ (Sigma-Aldrich, St. Louis, MO, USA) (density of 1.077–1.083 g/mL), Lymphoprep™ (Alere Technologies, Jena, Germany) (density of 1.077 g/mL), or Percoll™ (Cytiva, GE Healthcare, Uppsala, Sweden) (density of 1.070–1.080 g/mL). The choice of the appropriate gradient depends on the swine’s age, as this affects the efficiency of erythrocyte separation. For piglets, a gradient with a density of 1.075 g/mL is used, whereas for adult swine, a density of 1.077 g/mL is recommended [21]. After centrifugation in the density gradient medium, the cells undergo multiple washes in PBS (two or three times), which improves their viability and removes potential contaminants, including platelets [22]. In the case of bovine cells, washing steps help to remove residual platelets that might otherwise interfere with downstream analyses or cellular functions [23]. An alternative to the conventional methods utilizing standard tubes when applying blood onto a density gradient is the use of specialized test tubes equipped with separation inserts. These inserts serve as physical barriers that facilitate the formation of a distinct PBMC layer while minimizing contamination and improving the efficiency of the isolation process. Among these, SepMate™ (Stemcell Technologies, Vancouver, BC, Canada), visible in Figure 2, and Leucosep® (Greiner Bio-One, Kremsmünster, Austria) contain porous inserts that streamline cell separation by preventing the unwanted mixing of layers and reducing the need for precise pipetting, thereby enhancing reproducibility. In contrast, CPT™ (BD Vacutainer®, Becton, Dickinson and Company, Franklin Lakes, NJ, USA) offers a fully integrated system with a pre-filled density gradient medium, eliminating the need for additional reagents and manual gradient preparation, making it particularly suited for standardized clinical and high-throughput applications.
A comparison of human cell isolation methods has shown that CPT™ and SepMate™ are effective alternatives to the traditional Ficoll-Paque™ method, providing comparable cell quality and functionality while offering higher cell recovery [24,25]. Moreover, isolated PBMCs using the above tubes exhibit better functionality, particularly in terms of immune responses, increased cytokine production, improved proliferative capacity in response to stimulation, and greater efficiency in cytotoxic assays [24].

3. Cryopreservation of pPBMCs

After isolation, PBMCs often require storage for further research or laboratory analyses. Cryopreservation, which involves storing cells at extremely low temperatures, enables their long-term storage and subsequent use in immunological studies, such as analyzing the immune system’s response to infections, vaccines, or therapies. However, to preserve cell viability, phenotype, and functionality, the freezing process must be carried out in a controlled manner, minimizing the risk of damage caused by ice crystal formation or osmotic imbalances [24].

3.1. Preparation for Cryopreservation

A key step in preparing cells for freezing is the addition of cryoprotectants. The most commonly used agent is a mixture containing 10% dimethyl sulfoxide (DMSO) and 90% fetal bovine serum (FBS). Alternatively, ready-to-use cryopreservation media, such as PSC (Thermo Fisher Scientific, Waltham, MA, USA) or CryoStor CS10 (Stemcell Technologies, Vancouver, BC, Canada), which also rely on DMSO, are available. Studies indicate that reducing the DMSO concentration to 5% may improve human cell viability after thawing [26]. Furthermore, the comparisons of various cryoprotective media have shown no significant differences in cell survival rates, which remained above 89%. Functional tests, such as interferon gamma (IFN-γ) enzyme-linked immuno-spot ELISpot in response to phytohemagglutinin (PHA) and immunoglobulin G (IgG) ELISpot in response to the mitogenic cocktail resiquimod + interleukin-2 (R848 + IL-2), confirmed that results obtained using cryopreserved pPBMCs are comparable to those obtained using fresh cells [13]. A post-thaw viability of human peripheral blood mononuclear cells (hPBMCs) of 70–80% is considered sufficient for further analyses, minimizing the risk of experimental errors [27].
Cell concentration during freezing is crucial for their survival. A concentration that is too high can lead to mechanical damage caused by ice crystals, while a concentration that is too low reduces the protection provided by cryoprotectants. The recommended concentration is 106–107 PBMCs/mL, although the final choice depends on experimental requirements and the protocol used [28,29,30].

3.2. Control of the Freezing Rate

The cooling rate is another critical factor influencing the effectiveness of cryopreservation. Freezing too quickly can lead to osmotic imbalances, while freezing too slowly promotes the formation of large ice crystals, which can damage cells. According to the HIV/AIDS Network Coordination’s standard operating procedure (HANC-SOP) guidelines, the optimal freezing rate is −1 °C/min [31]. In practice, a two-step freezing method is used: in the first step, cells are gradually cooled in ultra-low temperature freezers (ULT) to approximately −80 °C, typically using a controlled-rate freezing container such as Mr. Frosty (manufactured by, e.g., Thermo Fisher Scientific, Waltham, MA, USA, which contains isopropanol to ensure a consistent cooling rate of ~1 °C per minute), and then transferred to the gas phase of liquid nitrogen (LN2), where the temperature drops below −150 °C (HANC-SOP). Storing cells at −80 °C for extended periods can lead to a decline in their viability and functionality, so it is recommended to transfer samples to liquid nitrogen as quickly as possible [32,33,34]. Although pPBMCs are highly sensitive to cryopreservation, long-term storage in liquid nitrogen remains the gold standard [35].

3.3. Thawing Process

Thawing is an equally important step in cryopreservation. The rapid thawing of cells in a 37 °C water bath for 2–5 min is recommended. After thawing, it is necessary to remove DMSO, which can be toxic to cells. For this purpose, cells are washed in culture medium by centrifugation at 300× g for 5 min and then resuspended in fresh medium [36,37].

4. Key Differences Between Human and Porcine PBMCs: Immunological Characteristics and Methodological Considerations

The isolation, characterization, and application of PBMCs differ between humans and swine due to both biological and methodological factors. Understanding these differences is essential for optimizing porcine PBMC (pPBMC) protocols and ensuring accurate data interpretation when comparing results across species.

4.1. Immunological and Phenotypic Differences

As of October 2024, the Human Cell Differentiation Molecules (HCDM) organization has assigned designation numbers up to CD371 for the human cluster of differentiation (CD) molecules, reflecting the ongoing advancement in the characterization of the human immune system [38]. In pigs, a landmark study conducted in 2018 identified 359 proteins homologous to human CD markers. Among these, 266 porcine CD molecules have specific antibody sets available, allowing for their detection and functional analysis [39]. Despite this progress, the comprehensive characterization of porcine CD molecules remains an active area of research.
One of the main differences between pPBMCs and hPBMCs is the presence of a higher number of double-positive T lymphocytes (CD4+CD8+) (Figure 3) in the peripheral blood of swine, which is not typical for human blood, where T cells are usually distinctly divided into CD4+ and CD8+ populations [40,41]. These porcine double-positive T cells express CD8αα, as opposed to the CD8αβ found on conventional cytotoxic T cells, and are functionally characterized as a memory T cell population with major histocompatibility complex class II (MHC-II) restriction [40]. A distinct population of circulating double-positive CD4+CD8αα+ αβ T cells was also identified in pigs. After integrating porcine single-cell RNA sequencing (scRNA-seq) data with human datasets, these porcine double positive T cell clusters showed high prediction scores, aligning with human CD4 central memory T cells (Tcm) [41].
In swine, especially young individuals, a higher number of γδ T cells is observed, whereas in human, Tαβ cells predominate. Specifically, minipigs, particularly juveniles, exhibit a higher proportion of T-cell receptor (TCR)-Tγδ+ cells compared to humans, where TCR-Tαβ+ cells are more prevalent [42]. NK cells in swine exhibit differences in surface marker expression compared to their human counterparts, which affects their cytotoxic function. In pigs, NK cells show variable expression of NKp46, which helps categorize them into functional subsets, NKp46, NKp46+, and NKp46high, based on their cytotoxic activity. Additionally, unlike human cells, porcine NK cells express NKp44 at high levels even without activation, with most NK cells expressing it ex vivo. This expression increases further upon stimulation [43,44].
Monocytes in both species share similar properties, though they differ in the expression of certain surface receptors. For example, porcine monocytes express higher levels of CD14 and lower levels of CD16 compared to human monocytes. Furthermore, one study identified two distinct subsets of porcine monocytes based on CD163 expression: CD14low/CD163+ and CD14hi/CD163 [45].
Additionally, responses to cytokines and chemokines may differ, influencing how PBMCs react to infections and inflammatory conditions. A study investigated how pPBMCs respond to stimulation with concanavalin A (Con A) [46]. They observed that the kinetics of cytokine messenger RNA (mRNA) expression differed between whole blood cultures and isolated PBMC cultures. For example, in PBMC cultures, peak mRNA levels occurred at 12 h for IL-2, 24 h for IL-6, 6 h for IL-8, and 12 h for IL-10. In contrast, whole blood cultures exhibited peak mRNA levels for these cytokines at 24 h for IL-2, 12 h for IL-6, 6 h for IL-8, and 24 h for IL-10. Furthermore, PBMC cultures consistently showed higher IL-2 mRNA levels, while whole blood cultures had higher IL-6 mRNA levels [46]. Genetic and evolutionary divergences also lead to the distinct regulation of immunology-related genes, such as those involved in cytokine production. A discovery highlighting species-specific regulatory mechanisms in porcine immune cells was made in a study analyzing how pPBMCs respond to lipopolysaccharide (LPS) and dexamethasone (DEX), revealing species-specific immune regulation. LPS increased the expression of pro-inflammatory cytokines through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation while also triggering anti-inflammatory mechanisms. DEX influenced immune responses by both inhibiting and activating different pathways, emphasizing the complex interaction between glucocorticoid signaling and immune regulation [47].
Despite these differences, pPBMCs exhibit many functional similarities to hPBMCs, making them a valuable model in biomedical research. Single-cell transcriptomics studies confirm both homologies and specific differences in gene expression between pPBMCs and hPBMCs. While many immune cell types, including T and B cells, monocytes, and dendritic cells, showed conserved gene expression patterns, notable species-specific differences were identified. In particular, γδ T cells and double-positive CD4+CD8+ T cells displayed distinct gene expression profiles in pigs [41,48]. The responses to immunological stimulation and immunosuppressive drugs were also analyzed, revealing similar T-cell activation mechanisms in both species. The mechanisms were similar between the two species, but notable differences existed in the activation levels of specific T-cell subsets. Specifically, human CD4+ T cells exhibited higher activation levels, whereas minipig CD8+ T cells were more abundant and showed greater activation. Additionally, the study found that the effectiveness of immunosuppressive drugs varied depending on the stimuli used, emphasizing the need to consider these differences when evaluating immunomodulatory drugs across species [42]. Understanding these relationships is crucial for fully leveraging the potential of pPBMCs as a model system.
Figure 3. Comparison of the most characteristic markers for porcine and human PBMCs. The bracket indicates that the identification of these cells requires the use of multiple markers, which allows for their accurate differentiation. Abbreviations: B = B lymphocyte; CD = cluster of differentiation; FoxP3 = Forkhead box protein P3; γδ T = gamma–delta T lymphocyte; mDC = myeloid dendritic cell; NK = natural killer cell; pDC = plasmacytoid dendritic cell; PBMC = peripheral blood mononuclear cells; T = T lymphocyte; Tc = cytotoxic T lymphocyte; TCR = T cell receptor; Th = T helper lymphocyte; Treg = regulatory T lymphocyte [39,49,50,51,52,53,54,55,56,57,58].
Figure 3. Comparison of the most characteristic markers for porcine and human PBMCs. The bracket indicates that the identification of these cells requires the use of multiple markers, which allows for their accurate differentiation. Abbreviations: B = B lymphocyte; CD = cluster of differentiation; FoxP3 = Forkhead box protein P3; γδ T = gamma–delta T lymphocyte; mDC = myeloid dendritic cell; NK = natural killer cell; pDC = plasmacytoid dendritic cell; PBMC = peripheral blood mononuclear cells; T = T lymphocyte; Tc = cytotoxic T lymphocyte; TCR = T cell receptor; Th = T helper lymphocyte; Treg = regulatory T lymphocyte [39,49,50,51,52,53,54,55,56,57,58].
Jcm 14 03432 g003

4.2. Methodological Considerations in pPBMC Isolation and Handling

Although separation and storage techniques for PBMCs may seem similar across species, the specific characteristics of porcine cells—resulting from differences in blood composition or immune system dynamics—require special attention. The isolation of porcine cells, while generally based on protocols used for hPBMCs, exhibits some differences, which are detailed in Table 1.
Heparin is more commonly used as an anticoagulant for porcine cells, consistent with recommendations for T-cell isolation [59,60]. Minor differences can also be observed in the dilution ratios of collected blood. Human blood is typically diluted 1:1 with PBS or Hank’s balanced salt solution (HBSS), while porcine blood is diluted 1:1 or 1:2, most often using PBS or RPMI 1640.
Regardless of the density gradient used, variability in centrifugation parameters is observed in protocols for porcine cells. Higher centrifugal acceleration (g) values are often used compared to standard procedures for hPBMCs. These differences likely arise from the distinct biological properties of the cells. The recommended temperature for both cell types is 20–25 °C, and the centrifugation time is typically around 30 min, which is commonly accepted for both swine and human.
The number of washes depends mainly on the degree of platelet contamination and the turbidity of the supernatant. For pPBMCs, 2–3 washes are typically performed, though centrifugation parameters can be difficult to standardize due to noticeable differences or a lack of precise guidelines in the literature.
Table 1. The isolation of PBMC from swine and human blood: a comparative analysis. Abbreviations: CD = citrate dextrose; EDTA = ethylenediaminetetraacetic acid; HBSS = Hank’s balanced salt solution; n.p. = not provided; PBS = phosphate-buffered saline; PLT = platelets; RPMI 1640 = Roswell Park Memorial Institute 1640 medium.
Table 1. The isolation of PBMC from swine and human blood: a comparative analysis. Abbreviations: CD = citrate dextrose; EDTA = ethylenediaminetetraacetic acid; HBSS = Hank’s balanced salt solution; n.p. = not provided; PBS = phosphate-buffered saline; PLT = platelets; RPMI 1640 = Roswell Park Memorial Institute 1640 medium.
SpeciesRef.Anti-
Coagulant
Ratio and DiluentDensity GradientCell Separation ParametersNumber of Washes and Washing SolutionWashing
Parameters
swine[61]EDTA1:2
PBS
Histopaque®-1077400× g
30 min; RT
2 washes,
PBS
250× g
10 min; RT
[62]heparin1:1
RPMI 1640
Histopaque®-10771100× g
25 min; RT
2 washes,
n.p.
n.p.
[63]heparinn.p.BD Vacutainer® CPT™500× g
30 min
3 washes,
PBS
n.p.
[64]heparin1:2
RPMI 1640
Histopaque®-10771100× g
25 min
2 washes,
RPMI 1640
n.p.
[65]n.p.1:1
PBS
Histopaque®-10772200 rpm
RT
3 washes,
PBS
2500 rpm
15 min; 4 °C
[66]heparinn.p.BD Vacutainer® CPT™1500× g
30 min; RT
2 washes,
PBS
700× g
10 min
[67]EDTA1:1
PBS + 20% CD
+ 2% FBS
SepMate™-50 tube with Lymphoprep™1100× g
10 min
until PLT removal, PBS + 20% ACD
+ 2% FBS
n.p.
[68]EDTA1:1
PBS
Ficoll-Paque™400× g
30 min; 25 °C
3 washes,
n.p.
1. 300× g
10 min; 4 °C;
2. 250× g
10 min; 4 °C
[69]heparin1:1
HBSS
Ficoll-Hypaque472× g
30 min; RT
1 wash,
HBSS
n.p.
[70]heparin2:1
PBS
Ficoll-Paque™ PLUS1455× g
30 min; RT
3 washes,
PBS
930× g
5 min; 4 °C
human[71]n.p.1:1
HBSS
Ficoll-Hypaque2000 r/min
20 min
2 washes,
HBSS
n.p.
[72]EDTA1:1
PBS
Ficoll-Paque™ PLUS400× g
30 min; 20 °C
1 wash,
PBS
300× g 10 min; 20 °C
[73]EDTA1:1
PBS
LSM™400× g
30 min
2 washes,
PBS
n.p.
[74]heparinn.p.Histopaque®-10772000 r/min
20 min; 4 °C
2 washes,
PBS
1500 r/min
10 min; 4 °C
[75]n.p.1:1
PBS + 2% FBS
Lymphoprep™800× g
20 min; RT
n.p.n.p.
[76]n.p.PBSLympholyte®1500 r/min
30 min; RT
n.p.
PBS
n.p.
[77]EDTA1:1
PBS
Histopaque®-1077800× g
30 min; 22 °C
1 wash,
PBS
300× g
3 min; 22 °C
[78]EDTA1:1
PBS
Ficoll-Paque™ PLUS400× g
30 min; 25 °C
2 washes,
PBS
300× g
10 min; 25 °C
[79]EDTA1:1
PBS + 2% FBS
Lymphoprep™800× g
20 min; RT
until the supernatant is clear, PBSn.p.
[80]sodium
citrate
1:1
PBS
Lymphoprep™1. 160× g
20 min; RT;
2. 800× g
20 min
3 washes,
RPMI
n.p.

5. Utilization of Porcine PBMCs

PBMCs are increasingly being used as a translational model in biomedical research, including preclinical safety assessments and drug development. This is due to their significant physiological, anatomical, and genetic similarities to human [42,81,82,83]. Swine are characterized by their organ sizes, metabolic processes, and functions of the cardiovascular, digestive, and immune systems that are comparable to those of humans, making them a more suitable model than smaller laboratory animals, such as rodents [82,84,85]. Additionally, the porcine genome shows greater similarity to the human genome, facilitating disease modeling and the testing of gene therapies. Swine are also widely used in toxicological, pharmacokinetic, and surgical studies, as well as in the testing of medical implants [81,85,86,87,88]. Despite challenges such as higher maintenance costs and ethical considerations, swine remain a valuable tool in biomedical research, contributing to the development of effective therapies for humans.

5.1. Immunological and Translational Applications of pPBMCs

Porcine PBMCs serve as a valuable model for dissecting fundamental immune mechanisms and testing therapeutic interventions, making them ideal for translational immunology research. These cells are widely used to study immune responses to various stimuli, providing insights into the mechanisms of innate and adaptive immunity. For instance, stimulation with LPS, a component of Gram-negative bacterial cell walls, induces the production of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), IL-1β, and IL-6 through activation of the toll-like receptor 4 (TLR4) pathway, closely mirroring responses in humans [15,89,90]. Similarly, polyinosinic:polycytidylic acid (Poly I:C), a viral double-stranded RNA mimic, activates the TLR3 pathway, leading to interferon production and antiviral gene expression [15].
Transcriptomic analyses using RNA sequencing (RNA-seq) have identified 603 genes specific to LPS stimulation, 254 to Poly I:C, and 882 to exposure to various microorganisms (EM), with 38 genes common across all three stimuli. Functional analyses show that LPS and Poly I:C activate pathways involved in inflammation and immune signaling, while EM exposure enhances metabolic responses, suggesting relevance for modeling sepsis, endotoxemia, and viral infections [15].
In vaccine development, pPBMCs play a crucial role in assessing immunological efficacy. Post-vaccination, the production of antigen-specific antibodies, T-cell proliferation, and cytokine secretion are analyzed [82,91,92,93,94,95,96,97]. Similarly, studies on influenza vaccines utilize PBMCs to assess cellular responses, which may contribute to the development of universal vaccines. These studies underscore the importance of PBMCs in optimizing vaccine formulations and understanding the mechanisms of immune memory.
Porcine PBMCs are also used to study the effects of immunomodulatory factors, which can enhance or suppress the immune response. For example, studies on enrofloxacin have shown that this antibiotic can transiently inhibit the production of pro-inflammatory cytokines in LPS-stimulated cells, suggesting immunomodulatory effects beyond its antibacterial properties [89]. Similarly, corticosteroids, commonly used to treat inflammatory conditions, inhibit cytokine production in PBMCs, providing a model for human therapy research [47,98].

5.2. Pathogen-Specific Infectious Disease Research Using pPBMCs

Porcine PBMCs are extensively used to investigate host–pathogen interactions in the context of viral and bacterial infections, especially those with zoonotic potential or significant impact on swine health. One of the most studied pathogens is the porcine reproductive and respiratory syndrome virus (PRRSV), which causes immunosuppression and chronic infection by modulating cytokine production and disrupting T-cell function [94,96,97].
Studies on vaccines against porcine reproductive and respiratory syndrome virus (PRRSV) have shown that the production of IFN-γ by virus-specific T cells correlates with protection against infection. A particularly important role is played by cytotoxic CD4+ T cells, which, upon in vitro restimulation, exhibit the increased expression of activation markers such as CD107a and IFN-γ, suggesting their critical role in protection against PRRSV-1 infection [99].
Another significant pathogen is the African swine fever virus (ASFV), characterized by high mortality rates. ASFV induces the production of pro-inflammatory cytokines and chemokines in pPBMCs, which correlates with disease severity [100]. Additionally, research on other viruses, such as classical swine fever virus (CSFV) and porcine circovirus type 2 (PCV2), utilizes pPBMCs to understand host–pathogen interactions and the kinetics of immune responses [101,102,103,104].
Porcine PBMCs are also a valuable tool in research on bacterial diseases, helping to elucidate the complex interactions between the host and pathogen. An example is Streptococcus suis, a zoonotic pathogen that causes meningitis in both swine and human. Infection with this pathogen induces both pro-inflammatory and anti-inflammatory cytokines in pPBMCs, with IL-10 playing a key role in modulating the inflammatory response [105,106]. Another important pathogen is Salmonella, particularly Salmonella typhimurium, which activates macrophages and dendritic cells in pPBMCs, initiating the host immune response [89]. These studies provide insights into the body’s defense mechanisms and potential therapeutic strategies.

5.3. Xenotransplantation

Transplantation, as a field of medicine focused on organ and tissue grafting, has long faced challenges such as donor shortages and graft rejection by the recipient’s immune system. In response to these challenges, scientists are increasingly developing xenotransplantation, where swine are considered promising donors due to the anatomical and physiological similarities of their organs—such as the heart and kidneys—to those of humans. However, the key obstacle remains the immunological barrier: human immune cells quickly recognize swine tissues as foreign, leading to rejection. In this context, pPBMCs play an indispensable role in research on immunological mechanisms, testing therapies, and enhancing cross-species compatibility.
Mixed porcine lymphocyte cultures with human cells are useful for assessing proliferative responses, indicating the potential risk of graft rejection [107,108,109]. The primary immunological barrier is the galactose-α-1,3-galactose (α-Gal) antigen, produced by the enzyme α-1,3-galactosyltransferase (GGTA1), which triggers natural antibody responses in humans. Genetic modifications, such as knockout of the GGTA1 gene, reduce α-Gal expression, thereby decreasing immunological reactivity [107,109,110,111,112,113,114,115,116].
Further genetic modifications, including the introduction of human complement regulatory proteins such as CD55 and CD59, are being tested in pig cells to enhance their resistance to complement-mediated lysis when exposed to human immune components. These modifications aim to address one of the major immunological barriers in xenotransplantation—hyperacute rejection—by inhibiting the activation and amplification of the complement cascade. Using hPBMCs as an in vitro model, researchers are evaluating the protective effects of these transgenes on porcine endothelial and other relevant cell types. The expression of CD55 interferes with the formation of C3/C5 convertases, while CD59 inhibits the formation of the membrane attack complex (MAC), both crucial mechanisms in mitigating complement-induced cytotoxicity. These approaches are being explored in combination with other genetic alterations, such as the deletion of α1,3-galactosyltransferase (GGTA1), to produce multi-transgenic pigs that are more compatible with the human immune system and potentially suitable for clinical xenotransplantation [107,109]. Additionally, PBMCs help elucidate the role of other antigen-presenting cells, such as porcine dendritic cells (DC research indicates that inflammatory signals, such as TNF-α, accelerate porcine DC maturation, enhancing their ability to activate human T cells and increasing the risk of graft rejection). This highlights the importance of controlling inflammation to reduce immune activation [117]. Conversely, transforming growth factor β1 (TGF-β1) has been shown to influence the differentiation of porcine monocytes into Langerhans cell-like dendritic cells (MoLCs), a specialized subset of antigen-presenting cells. These porcine MoLCs exhibit distinct surface markers and functional properties, including high expression of major histocompatibility complex (MHC) molecules and co-stimulatory proteins, which enhance their capacity to present antigens and activate T cells. Their potent immunostimulatory function positions them as key contributors to immune surveillance and rejection processes in xenotransplantation [109]. These advancements, tested using pPBMCs, contribute to reducing the risk of graft rejection.
pPBMCs are a valuable in vitro model for evaluating the efficacy of immunosuppressive drugs in xenotransplantation research. One such compound, rapamycin, has been shown to significantly inhibit pPBMC proliferation in response to mitogenic stimuli, such as concanavalin A (ConA) or phytohemagglutinin (PHA), by interfering with the mechanistic target of rapamycin (mTOR) signaling pathway. This suppression helps mitigate immune activation that can lead to graft rejection. Notably, studies examining pPBMCs restimulated with porcine circovirus type 2 (PCV2) antigen—a model for antigen-specific immune responses—demonstrated that rapamycin effectively reduces antigen-driven lymphocyte proliferation as well. These findings emphasize that the efficacy of immunosuppressive agents like rapamycin is not uniform but highly dependent on the nature of the immune challenge. As a result, tailoring immunosuppressive therapy to match the specific immunological context—whether it involves alloantigen, xenoantigen, or viral antigen stimulation—is crucial for optimizing graft acceptance while minimizing immune suppression-related complications [42].
Strategies for inducing tolerance, such as hematopoietic stem cell transplantation or the expression of immune-inhibitory molecules, are being intensively explored using pPBMC models [118,119,120]. Monitoring immune responses post-transplantation using pPBMCs allows for the identification of early signs of rejection or tolerance, which is crucial for clinical applications [97,110,112,121].

6. Conclusions

Porcine PBMCs have emerged as a critical tool in modern biomedical research, offering a robust, reproducible, and physiologically relevant model for studying immune function, disease mechanisms, and therapeutic responses. The isolation and preservation techniques described herein ensure high cell viability and functional integrity, allowing for a broad range of experimental applications. The unique immunophenotypic characteristics of pPBMCs, including the presence of CD4+CD8+ T cells and species-specific cytokine expression patterns, provide both opportunities and challenges when extrapolating findings to human systems. However, comparative studies using advanced transcriptomic and functional analyses have revealed a considerable overlap in immune responses between porcine and human PBMCs. This reinforces the utility of swine as a translational model, particularly in fields such as vaccine development, immunomodulatory therapy, infectious disease research, and xenotransplantation. Advances in in vitro systems utilizing pPBMCs have significantly expanded their potential for use in preclinical studies, particularly where human samples are limited or ethically restricted.
As the demand for reliable large-animal models grows, the porcine immune system—especially through pPBMC-based approaches—offers a practical and physiologically relevant platform for bridging basic research with clinical applications. Thanks to their anatomical, physiological, and immunogenetic similarities to humans, porcine PBMCs are becoming an increasingly important tool in translational medicine.

Author Contributions

Conceptualization, M.P., M.Z.-W. and M.C.-M.; funding acquisition, M.Z.-W.; investigation, M.P.; project administration, M.P. and M.Z.-W.; supervision, M.Z.-W.; visualization, M.P. and M.Z.-W.; writing—original draft preparation, M.P. and M.Z.-W.; writing—review and editing, M.C.-M. and M.Z.-W. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by the Polish Ministry of Health subvention according to the number SUBK.A020.24.083 (to M.Z.-W.) from the IT Simple system from Wroclaw Medical University, Wroclaw, Poland.

Acknowledgments

All figures were created with BioRender.com, Figure 1—Pietrzak, M. (2025) https://BioRender.com/ek7zg6j (accessed on 10 May 2025); Figure 2https://BioRender.com/qaqhkq7 (accessed on 10 May 2025); Figure 3https://BioRender.com/qaqhkq7 (accessed on 10 May 2025). Magdalena Pietrzak is a scholarship recipient working under the NCN-OPUS grant at Wroclaw Medical University.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
α-Galalpha-galactosidase
γδ Tgamma–delta T cell
ASFVAfrican Swine Fever Virus
BB lymphocyte
CDcluster of differentiation
DCdendritic cell
DEXdexamethasone
DMSOdimethyl sulfoxide
EDTAethylenediaminetetraacetic acid
ELISpotenzyme-linked ImmunoSpot
FBSfetal bovine serum
GGTA1gene encoding alpha1,3-galactosyltransferase
HANC-SOPHIV/AIDS Network Coordination standard operating procedures
HBSSHank’s balanced salt solution
hPBMChuman peripheral blood mononuclear cell
IFN-γinterferon gamma
ILinterleukin
LN2liquid nitrogen
LPSlipopolysaccharide
mRNAmessenger RNA
MHCmajor histocompatibility complex class
NKnatural killer cell
NKpnatural killer protein
n.p.not provided
PBMCperipheral blood mononuclear cell
PBSphosphate-buffered saline
PCV2porcine circovirus type 2
PolyI:Cpolyinosinic:polycytidylic acid
pPBMCporcine peripheral blood mononuclear cell
PRRSVporcine reproductive and respiratory syndrome virus
RBCred blood cell
RPMI 1640Roswell Park Memorial Institute 1640 (cell culture medium)
RTroom temperature
Tccytotoxic T cell
ThT helper cell
TLRtoll-like receptor
TNF-αtumor necrosis factor alpha
Tregregulatory T cells

References

  1. Gerner, W.; Käser, T.; Saalmüller, A. Porcine T lymphocytes and NK cells—An update. Dev. Comp. Immunol. 2009, 33, 310–320. [Google Scholar] [CrossRef]
  2. Cunha, P.; Gilbert, F.B.; Bodin, J.; Godry, L.; Germon, P.; Holbert, S.; Martins, R.P. Simplified Approaches for the Production of Monocyte-Derived Dendritic Cells and Study of Antigen Presentation in Bovine. Front. Vet. Sci. 2022, 9, 891893. [Google Scholar] [CrossRef] [PubMed]
  3. Wojciechowska-Durczynska, K.; Wieczorek-Szukala, K.; Stefanski, B.; Zygmunt, A.; Stepniak, J.; Karbownik-Lewinska, M.; Lewinski, A. Percentage of Myeloid Dendritic Cells in Peripheral Venous Blood Is Negatively Related to Incidence of Graves’ Orbitopathy. Mediat. Inflamm. 2021, 2021, 8896055. [Google Scholar] [CrossRef]
  4. Larsson, A.M.; Nordström, O.; Johansson, A.; Rydén, L.; Leandersson, K.; Bergenfelz, C. Peripheral Blood Mononuclear Cell Populations Correlate with Outcome in Patients with Metastatic Breast Cancer. Cells 2022, 11, 1639. [Google Scholar] [CrossRef] [PubMed]
  5. Le Page, L.; Baldwin, C.L.; Telfer, J.C. γδ T cells in artiodactyls: Focus on swine. Dev. Comp. Immunol. 2022, 128, 104334. [Google Scholar] [CrossRef]
  6. Pabst, R. The pig as a model for immunology research. Cell Tissue Res. 2020, 380, 287–304. [Google Scholar] [CrossRef] [PubMed]
  7. Lunney, J.K.; Van Goor, A.; Walker, K.E.; Hailstock, T.; Franklin, J.; Dai, C. Importance of the pig as a human biomedical model. Sci. Transl. Med. 2021, 13, eabd5758. [Google Scholar] [CrossRef]
  8. Mair, K.H.; Sedlak, C.; Käser, T.; Pasternak, A.; Levast, B.; Gerner, W.; Saalmüller, A.; Summerfield, A.; Gerdts, V.; Wilson, H.L.; et al. The porcine innate immune system: An update. Dev. Comp. Immunol. 2014, 45, 321–343. [Google Scholar] [CrossRef]
  9. Sipos, W. Shifts in porcine PBMC populations from adolescence to adulthood. Vet. Immunol. Immunopathol. 2019, 211, 35–37. [Google Scholar] [CrossRef]
  10. Mattoo Sul, S.; Aganja, R.P.; Kim, S.C.; Jeong, C.G.; Nazki, S.; Khatun, A.; Kim, W.I.; Lee, S.M. A standardized method to study immune responses using porcine whole blood. J. Vet. Sci. 2023, 24, 1–14. [Google Scholar] [CrossRef]
  11. Overgaard, N.H.; Frøsig, T.M.; Welner, S.; Rasmussen, M.; Ilsøe, M.; Sørensen, M.R.; Andersen, M.H.; Buus, S.; Jungersen, G. Establishing the pig as a large animal model for vaccine development against human cancer. Front. Genet. 2015, 6, 286. [Google Scholar] [CrossRef] [PubMed]
  12. Díaz, I. Rules of thumb to obtain, isolate, and preserve porcine peripheral blood mononuclear cells. Vet. Immunol. Immunopathol. 2022, 251, 110461. [Google Scholar] [CrossRef] [PubMed]
  13. Li, Y.; Mateu, E.; Díaz, I. Impact of Cryopreservation on Viability, Phenotype, and Functionality of Porcine PBMC. Front. Immunol. 2021, 12, 765667. [Google Scholar] [CrossRef]
  14. Cao, Q.M.; Ni, Y.Y.; Cao, D.; Tian, D.; Yugo, D.M.; Heffron, C.L.; Overend, C. Recombinant Porcine Reproductive and Respiratory Syndrome Virus Expressing Membrane-Bound Interleukin-15 as an Immunomodulatory Adjuvant Enhances NK and γδ T Cell Responses and Confers Heterologous Protection. J. Virol. 2018, 92, 10–1128. [Google Scholar] [CrossRef]
  15. Yang, J.; Chen, S.; Ma, F.; Ding, N.; Mi, S.; Zhao, Q.; Xing, Y.; Yang, T.; Xing, K.; Yu, Y.; et al. Pathogen stimulations and immune cells synergistically affect the gene expression profile characteristics of porcine peripheral blood mononuclear cells. BMC Genom. 2024, 25, 719. [Google Scholar] [CrossRef]
  16. Mu, S.; Chen, L.; Dong, H.; Li, S.; Zhang, Y.; Yin, S.; Tian, Y.; Ding, Y.; Sun, S.; Shang, S.; et al. Enhanced antigen-specific CD8 T cells contribute to early protection against FMDV through swine DC vaccination. J. Virol. 2024, 98, e02002-23. [Google Scholar] [CrossRef]
  17. Hernandez-Franco, J.F.; Xie, S.; Thimmapuram, J.; Ragland, D.; HogenEsch, H. Mechanism of activation of porcine dendritic cells by an α-D-glucan nanoparticle adjuvant and a nanoparticle/poly(I:C) combination adjuvant. Front. Immunol. 2022, 13, 990900. [Google Scholar] [CrossRef] [PubMed]
  18. Khatri, M.; O’Brien, T.D.; Chattha, K.S.; Saif, L.J. Porcine lung mesenchymal stromal cells possess differentiation and immunoregulatory properties. Stem Cell Res. Ther. 2015, 6, 1–11. [Google Scholar] [CrossRef]
  19. Kapetanovic, R.; Fairbairn, L.; Beraldi, D.; Sester, D.P.; Archibald, A.L.; Tuggle, C.K.; Hume, D.A. Pig Bone Marrow-Derived Macrophages Resemble Human Macrophages in Their Response to Bacterial Lipopolysaccharide. J. Immunol. 2012, 188, 3382–3394. [Google Scholar] [CrossRef]
  20. Duvigneau, J.C.; Sipos, W.; Hartl, R.T.; Bayer, M.; Moldzio, R.; Stevenson, L.; Adair, B.; Gemeiner, M. Heparin and EDTA as anticoagulant differentially affect cytokine mRNA level of cultured porcine blood cells. J. Immunol. Methods 2007, 324, 38–47. [Google Scholar] [CrossRef]
  21. Talker, S.C.; Käser, T.; Reutner, K.; Sedlak, C.; Mair, K.H.; Koinig, H.; Graage, R.; Viehmann, M.; Klingler, E.; Ladinig, A.; et al. Phenotypic maturation of porcine NK- and T-cell subsets. Dev. Comp. Immunol. 2013, 40, 51–68. [Google Scholar] [CrossRef] [PubMed]
  22. Berg, C.; Wilker, S.; Roider, J.; Klettner, A. Isolation of porcine monocyte population: A simple and efficient method. Vet. Res. Commun. 2013, 37, 239–241. [Google Scholar] [CrossRef]
  23. Stabel, J.R.; Wherry, T.L.T. Comparison of methods to isolate peripheral blood mononuclear cells from cattle blood. J. Immunol. Methods 2023, 512, 113407. [Google Scholar] [CrossRef]
  24. Grievink, H.W.; Luisman, T.; Kluft, C.; Moerland, M.; Malone, K.E. Comparison of Three Isolation Techniques for Human Peripheral Blood Mononuclear Cells: Cell Recovery and Viability, Population Composition, and Cell Functionality. Biopreserv. Biobank. 2016, 14, 410–415. [Google Scholar] [CrossRef] [PubMed]
  25. Ruitenberg, J.J.; Mulder, C.B.; Maino, V.C.; Landay, A.L.; Ghanekar, S.A. VACUTAINER® CPTTM and Ficoll density gradient separation perform equivalently in maintaining the quality and function of PBMC from HIV seropositive blood samples. BMC Immunol. 2006, 7, 11. [Google Scholar] [CrossRef] [PubMed]
  26. Kaiser, D.; Otto, N.M.; McCallion, O.; Hoffmann, H.; Zarrinrad, G.; Stein, M.; Beier, C.; Matz, I.; Herschel, M.; Hester, J.; et al. Freezing Medium Containing 5% DMSO Enhances the Cell Viability and Recovery Rate After Cryopreservation of Regulatory T Cell Products ex vivo and in vivo. Front. Cell Dev. Biol. 2021, 9, 750286. [Google Scholar] [CrossRef]
  27. Betsou, F.; Gaignaux, A.; Ammerlaan, W.; Norris, P.J.; Stone, M. Biospecimen Science of Blood for Peripheral Blood Mononuclear Cell (PBMC) Functional Applications. Curr. Pathobiol. Rep. 2019, 7, 17–27. [Google Scholar] [CrossRef]
  28. Browne, D.J.; Miller, C.M.; Doolan, D.L. Technical pitfalls when collecting, cryopreserving, thawing, and stimulating human T-cells. Front. Immunol. 2024, 15, 1382192. [Google Scholar] [CrossRef]
  29. Hope, C.M.; Huynh, D.; Wong, Y.Y.; Oakey, H.; Perkins, G.B.; Nguyen, T.; Binkowski, S.; Bui, M.; Choo, A.Y.L.; Gibson, E.; et al. Optimization of blood handling and peripheral blood mononuclear cell cryopreservation of low cell number samples. Int. J. Mol. Sci. 2021, 22, 9129. [Google Scholar] [CrossRef]
  30. Weinberg, A.; Song, L.Y.; Wilkening, C.L.; Fenton, T.; Hural, J.; Louzao, R.; Ferrari, G.; Etter, P.E.; Berrong, M.; Canniff, J.D.; et al. Optimization of storage and shipment of cryopreserved peripheral blood mononuclear cells from HIV-infected and uninfected individuals for ELISPOT assays. J. Immunol. Methods 2010, 363, 42–50. [Google Scholar] [CrossRef]
  31. Cross-Network PBMC Processing SOP. Available online: https://www.hanc.info/resources/sops-guidelines-resources/laboratory/cross-network-pbmc-processing-sop.html (accessed on 10 May 2025).
  32. Hønge, B.L.; Petersen, M.S.; Olesen, R.; Møller, B.K.; Erikstrup, C. Optimizing recovery of frozen human peripheral blood mononuclear cells for flow cytometry. PLoS ONE 2017, 12, e0187440. [Google Scholar] [CrossRef] [PubMed]
  33. Weinberg, A.; Song, L.Y.; Wilkening, C.; Sevin, A.; Blais, B.; Louzao, R.; Stein, D. Optimization and limitations of use of cryopreserved peripheral blood mononuclear cells for functional and phenotypic T-cell characterization. CVI 2009, 16, 1176–1186. [Google Scholar] [CrossRef]
  34. Yang, J.; Diaz, N.; Adelsberger, J.; Zhou, X.; Stevens, R.; Rupert, A.; Metcalf, J.A.; Baseler, M.; Barbon, C.; Imamichi, T.; et al. The effects of storage temperature on PBMC gene expression. BMC Immunol. 2016, 17, 6. [Google Scholar] [CrossRef] [PubMed]
  35. Koch, E.; Larak, M.; Ellendorff, F. Comparative studies on in vitro reactivity of fresh and cryopreserved pig lymphocytes. Cryobiology 1991, 28, 405–412. [Google Scholar] [CrossRef]
  36. Cryopreserved PBMCs Retain Phenotype and Function. Available online: https://www.sigmaaldrich.com/PL/en/technical-documents/technical-article/cell-culture-and-cell-culture-analysis/primary-cell-culture/cryopreserved-pbmcs-retain-phenotype-and-function (accessed on 10 May 2025).
  37. Thawing Cells—Cell Culture Protocols. Available online: https://www.thermofisher.com/pl/en/home/references/gibco-cell-culture-basics/cell-culture-protocols/thawing-cells.html (accessed on 10 May 2025).
  38. New CD Molecules. Available online: https://www.hcdm.org/index.php/hlda10-workshop/new-cd-molecules (accessed on 10 May 2025).
  39. Dawson, H.D.; Lunney, J.K. Porcine cluster of differentiation (CD) markers 2018 update. Res. Vet. Sci. 2018, 118, 199–246. [Google Scholar] [CrossRef]
  40. Corbett, R.J.; Luttman, A.M.; Herrera-Uribe, J.; Liu, H.; Raney, N.E.; Grabowski, J.M.; Loving, C.L.; Tuggle, C.K.; Ernst, C.W. Assessment of DNA methylation in porcine immune cells reveals novel regulatory elements associated with cell-specific gene expression and immune capacity traits. BMC Genom. 2022, 23, 575. [Google Scholar] [CrossRef]
  41. Loving, C.L.; Wiarda, J.E.; Sivasankaran, S.K.; Daharsh, L.; Liu, H.; Byrne, K.A.; Tuggle, C.K. Characterization of circulating porcine immune cells using single-cell RNA-sequencing and comparison to human datasets. J. Immunol. 2021, 206, 19.17. [Google Scholar] [CrossRef]
  42. Pernold, C.P.S.; Lagumdzic, E.; Stadler, M.; Dolezal, M.; Jäckel, S.; Schmitt, M.W.; Mair, K.H.; Saalmüller, A. Species comparison: Human and minipig PBMC reactivity under the influence of immunomodulating compounds in vitro. Front. Immunol. 2024, 14, 1327776. [Google Scholar] [CrossRef] [PubMed]
  43. Mair, K.H.; Crossman, A.J.; Wagner, B.; Babasyan, S.; Noronha, L.; Boyd, P.; Zarlenga, D.; Stadler, M.; van Dongen, K.A.; Gerner, W.; et al. The Natural Cytotoxicity Receptor NKp44 (NCR2, CD336) Is Expressed on the Majority of Porcine NK Cells Ex Vivo Without Stimulation. Front. Immunol. 2022, 13, 767530. [Google Scholar] [CrossRef]
  44. Mair, K.H.; Essler, S.E.; Patzl, M.; Storset, A.K.; Saalmüller, A.; Gerner, W. NKp46 expression discriminates porcine NK cells with different functional properties. Eur. J. Immunol. 2012, 42, 1261–1271. [Google Scholar] [CrossRef]
  45. Fairbairn, L.; Kapetanovic, R.; Beraldi, D.; Sester, D.P.; Tuggle, C.K.; Archibald, A.L.; Hume, D.A. Comparative Analysis of Monocyte Subsets in the Pig. J. Immunol. 2013, 190, 6389–6396. [Google Scholar] [CrossRef] [PubMed]
  46. Yancy, H.; Ayers, S.L.; Farrell, D.E.; Day, A.; Myers, M.J. Differential cytokine mRNA expression in swine whole blood and peripheral blood mononuclear cell cultures. Vet. Immunol. Immunopathol. 2001, 79, 41–52. [Google Scholar] [CrossRef] [PubMed]
  47. Li, Z.; Trakooljul, N.; Hadlich, F.; Ponsuksili, S.; Wimmers, K.; Murani, E. Transcriptome analysis of porcine PBMCs reveals lipopolysaccharide-induced immunomodulatory responses and crosstalk of immune and glucocorticoid receptor signaling. Virulence 2021, 12, 1808–1824. [Google Scholar] [CrossRef]
  48. Herrera-Uribe, J.; Wiarda, J.E.; Sivasankaran, S.K.; Daharsh, L.; Liu, H.; Byrne, K.A.; Smith, T.P.L.; Lunney, J.K.; Loving, C.L.; Tuggle, C.K. Reference Transcriptomes of Porcine Peripheral Immune Cells Created Through Bulk and Single-Cell RNA Sequencing. Front. Genet. 2021, 12, 689406. [Google Scholar] [CrossRef] [PubMed]
  49. Tedder, T.F. CD19: A promising B cell target for rheumatoid arthritis. Nat. Rev. Rheumatol. 2009, 5, 572–577. [Google Scholar] [CrossRef]
  50. Mycko, M.P. The mechanism of action of anti-CD20 monoclonal antibodies used in the treatment of multiple sclerosis. Curr. Neurol. 2023, 23, 72–78. [Google Scholar] [CrossRef]
  51. Cooper, M.A.; Fehniger, T.A.; Caligiuri, M.A. The biology of human natural killer-cell subsets. Trends Immunol. 2001, 22, 633–640. [Google Scholar] [CrossRef]
  52. Ebner, F.; Schwiertz, P.; Steinfelder, S.; Pieper, R.; Zentek, J.; Schütze, N.; Baums, C.G.; Alber, G.; Geldhof, P.; Hartmann, S. Pathogen-Reactive T Helper Cell Analysis in the Pig. Front. Immunol. 2017, 8, 565. [Google Scholar] [CrossRef]
  53. Hoog, A.; Villanueva-Hernández, S.; Razavi, M.A.; van Dongen, K.; Eder, T.; Piney, L.; Chapat, L.; de Luca, K.; Grebien, F.; Mair, K.H.; et al. Identification of CD4+ T cells with T follicular helper cell characteristics in the pig. Dev. Comp. Immunol. 2022, 134, 104462. [Google Scholar] [CrossRef]
  54. Käser, T.; Gerner, W.; Hammer, S.E.; Patzl, M.; Saalmüller, A. Phenotypic and functional characterisation of porcine CD4+CD25high regulatory T cells. Vet. Immunol. Immunopathol. 2008, 122, 153–158. [Google Scholar] [CrossRef]
  55. Auray, G.; Keller, I.; Python, S.; Gerber, M.; Bruggmann, R.; Ruggli, N.; Summerfield, A. Characterization and Transcriptomic Analysis of Porcine Blood Conventional and Plasmacytoid Dendritic Cells Reveals Striking Species-Specific Differences. J. Immunol. 2016, 197, 4791–4806. [Google Scholar] [CrossRef] [PubMed]
  56. Feng, F.; Li, Z.; Xie, Q.; Song, W. Phenotypic and functional differences of dendritic cells in tumor. J. Cancer Res. Ther. 2023, 19, 1509–1516. [Google Scholar] [CrossRef]
  57. Summerfield, A.; McCullough, K.C. The porcine dendritic cell family. Dev. Comp. Immunol. 2009, 33, 299–309. [Google Scholar] [CrossRef] [PubMed]
  58. Porcine Antibodies—Innate Immunity. Available online: https://www.bio-rad-antibodies.com/porcine-antibodies-innate-immunity.html (accessed on 10 May 2025).
  59. Mallone, R.; Mannering, S.I.; Brooks-Worrell, B.M.; Durinovic-Belló, I.; Cilio, C.M.; Wong, F.S.; Schloot, N.C. Isolation and preservation of peripheral blood mononuclear cells for analysis of islet antigen-reactive T cell responses: Position statement of the T-Cell Workshop Committee of the Immunology of Diabetes Society. Clin. Exp. Immunol. 2010, 163, 33–49. [Google Scholar] [CrossRef]
  60. Scheible, K.; Secor-Socha, S.; Wightman, T.; Wang, H.; Mariani, T.J.; Topham, D.J.; Pryhuber, G.; Quataert, S. Stability of T cell phenotype and functional assays following heparinized umbilical cord blood collection. Cytom. Part A 2012, 81, 937–949. [Google Scholar] [CrossRef] [PubMed]
  61. Dozois, C.M.; Oswald, E.; Gautier, N.; Serthelon, J.P.; Fairbrother, J.M.; Oswald, I.P. A reverse transcription-polymerase chain reaction method to analyze porcine cytokine gene expression. Vet. Immunol. Immunopathol. 1997, 58, 287–300. [Google Scholar] [CrossRef]
  62. Begni, B.; Amadori, M.; Ritelli, M.; Podavini, D. Effects of IFN-α on the Inflammatory Response of Swine Leukocytes to Bacterial Endotoxin. JICR 2005, 25, 202–208. [Google Scholar] [CrossRef]
  63. Wikström, F.H.; Meehan, B.M.; Berg, M.; Timmusk, S.; Elving, J.; Fuxler, L.; Magnusson, M.; Allan, G.M.; McNeilly, F.; Fossum, C. Structure-Dependent Modulation of Alpha Interferon Production by Porcine Circovirus 2 Oligodeoxyribonucleotide and CpG DNAs in Porcine Peripheral Blood Mononuclear Cells. J. Virol. 2007, 81, 4919–4927. [Google Scholar] [CrossRef]
  64. Amadori, M.; Cristiano, A.; Ferrari, M. Constitutive expression of interferons in swine leukocytes. Res. Vet. Sci. 2010, 88, 64–71. [Google Scholar] [CrossRef]
  65. Sah, V.; Kumar, A.; Dhar, P.; Upmanyu, V.; Tiwari, A.K.; Wani, S.A.; Sahu, A.R.; Kumar, A.; Badasara, S.K.; Pandey, A.; et al. Signature of genome wide gene expression in classical swine fever virus infected macrophages and PBMCs of indigenous vis-a-vis crossbred pigs. Gene 2020, 731, 144356. [Google Scholar] [CrossRef]
  66. Platt, R.; Vincent, A.L.; Gauger, P.C.; Loving, C.L.; Zanella, E.L.; Lager, K.M.; Kehrli, M.E., Jr.; Kimura, K.; Roth, J.A. Comparison of humoral and cellular immune responses to inactivated swine influenza virus vaccine in weaned pigs. Vet. Immunol. Immunopathol. 2011, 142, 252–257. [Google Scholar] [CrossRef] [PubMed]
  67. Chung, C.J.; Cha, S.H.; Grimm, A.L.; Ajithdoss, D.; Rzepka, J.; Chung, G.; Yu, J.; Davis, W.C.; Ho, C.S. Pigs that recover from porcine reproduction and respiratory syndrome virus infection develop cytotoxic CD4+CD8+ and CD4+CD8- T-cells that kill virus infected cells. PLoS ONE 2018, 13, e0203482. [Google Scholar] [CrossRef] [PubMed]
  68. McCullough, K.C.; Schaffner, R.; Fraefel, W.; Kihm, U. The relative density of CD44-positive porcine monocytic cell populations varies between isolations and upon culture and influences susceptibility to infection by African swine fever virus. Immunol. Lett. 1993, 37, 83–90. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, J.; Xu, X.; Huang, X.; Zhu, H.; Chen, H.; Wang, W.; Liu, Y. Analysis of microRNA expression profiles in porcine PBMCs after LPS stimulation. Innate Immunol. 2020, 26, 435–446. [Google Scholar] [CrossRef]
  70. Franzoni, G.; Kurkure, N.V.; Essler, S.E.; Pedrera, M.; Everett, H.E.; Bodman-Smith, K.B.; Crooke, H.R.; Graham, S.P. Proteome-Wide Screening Reveals Immunodominance in the CD8 T Cell Response against Classical Swine Fever Virus with Antigen-Specificity Dependent on MHC Class I Haplotype Expression. PLoS ONE 2013, 8, 84246. [Google Scholar] [CrossRef]
  71. Shui, X.; Chen, S.; Lin, J.; Kong, J.; Zhou, C.; Wu, J. Knockdown of lncRNA NEAT1 inhibits Th17/CD4 + T cell differentiation through reducing the STAT3 protein level. J. Cell Physiol. 2019, 234, 22477–22484. [Google Scholar] [CrossRef]
  72. Costa, A.; Reynés, B.; Konieczna, J.; Martín, M.; Fiol, M.; Palou, A.; Romaguera, D.; Oliver, P. Use of human PBMC to analyse the impact of obesity on lipid metabolism and metabolic status: A proof-of-concept pilot study. Sci. Rep. 2021, 11, 18329. [Google Scholar] [CrossRef]
  73. Madden, L.A.; Vince, R.V.; Laden, G. The effect of acute hyperoxia in vivo on NF kappa B expression in human PBMC. Cell Biochem. Funct. 2011, 29, 71–73. [Google Scholar] [CrossRef]
  74. Chen, R.; Curran, J.; Pu, F.; Zhuola, Z.; Bayon, Y.; Hunt, J. In Vitro Response of Human Peripheral Blood Mononuclear Cells (PBMC) to Collagen Films Treated with Cold Plasma. Polymers 2017, 9, 254. [Google Scholar] [CrossRef]
  75. Robert, J.; Button, E.B.; Stukas, S.; Boyce, G.K.; Gibbs, E.; Cowan, C.M.; Gilmour, M.; Cheng, W.H.; Soo, S.K.; Yuen, B.; et al. High-density lipoproteins suppress Aβ-induced PBMC adhesion to human endothelial cells in bioengineered vessels and in monoculture. Mol. Neurodegener. 2017, 12, 1–19. [Google Scholar] [CrossRef]
  76. Steller, D.; Scheibert, A.; Sturmheit, T.; Hakim, S.G. Establishment and validation of an in vitro co-culture model for oral cell lines using human PBMC-derived osteoclasts, osteoblasts, fibroblasts and keratinocytes. Sci. Rep. 2020, 10, 16861. [Google Scholar] [CrossRef] [PubMed]
  77. Salustri, A.; De Maio, F.; Palmieri, V.; Santarelli, G.; Palucci, I.; Mercedes Bianco, D.; Marchionni, F.; Bellesi, S.; Ciasca, G.; Perini, G.; et al. Evaluation of the Toxic Activity of the Graphene Oxide in the Ex Vivo Model of Human PBMC Infection with Mycobacterium tuberculosis. Microorganisms 2023, 11, 554. [Google Scholar] [CrossRef] [PubMed]
  78. Cifre, M.; Díaz-Rúa, R.; Varela-Calviño, R.; Reynés, B.; Pericás-Beltrán, J.; Palou, A.; Oliver, P. Human peripheral blood mononuclear cell in vitro system to test the efficacy of food bioactive compounds: Effects of polyunsaturated fatty acids and their relation with BMI. Mol. Nutr. Food. Res. 2017, 61, 1600353. [Google Scholar] [CrossRef]
  79. Luukkainen, A.; Puan, K.J.; Yusof, N.; Lee, B.; Tan, K.S.; Liu, J.; Yan, Y.; Toppila-Salmi, S.; Renkonen, R.; Chow, V.T.; et al. A Co-culture Model of PBMC and Stem Cell Derived Human Nasal Epithelium Reveals Rapid Activation of NK and Innate T Cells Upon Influenza A Virus Infection of the Nasal Epithelium. Front. Immunol. 2018, 9, 2514. [Google Scholar] [CrossRef]
  80. Kaya, E.; Grassi, L.; Benedetti, A.; Maisetta, G.; Pileggi, C.; Di Luca, M.; Batoni, G.; Esin, S. In vitro Interaction of Pseudomonas aeruginosa Biofilms with Human Peripheral Blood Mononuclear Cells. Front. Cell Infect. Microbiol. 2020, 10, 187. [Google Scholar] [CrossRef]
  81. Bode, G.; Clausing, P.; Gervais, F.; Loegsted, J.; Luft, J.; Nogues, V.; Sims, J. The utility of the minipig as an animal model in regulatory toxicology. J. Pharmacol. Toxicol. Methods 2010, 62, 196–220. [Google Scholar] [CrossRef]
  82. Lunney, J.K. Advances in Swine Biomedical Model Genomics. Int. J. Biol. Sci. 2007, 3, 179–184. [Google Scholar] [CrossRef]
  83. Vodička, P.; Smetana, K.; Dvořánková, B.; Emerick, T.; Xu, Y.; Ourednik, J.; Ourednik, V.; Motlík, J. The Miniature Pig as an Animal Model in Biomedical Research. Ann. N. Y. Acad. Sci. 2005, 1049, 161–171. [Google Scholar] [CrossRef] [PubMed]
  84. Helke, K.L.; Swindle, M.M. Animal models of toxicology testing: The role of pigs. Expert Opin. Drug. Metab. Toxicol. 2013, 9, 127–139. [Google Scholar] [CrossRef]
  85. Swindle, M.M.; Makin, A.; Herron, A.J.; Clubb, F.J.; Frazier, K.S. Swine as Models in Biomedical Research and Toxicology Testing. Vet. Pathol. 2012, 49, 344–356. [Google Scholar] [CrossRef]
  86. Aigner, B.; Renner, S.; Kessler, B.; Klymiuk, N.; Kurome, M.; Wünsch, A.; Wolf, E. Transgenic pigs as models for translational biomedical research. J. Mol. Med. 2010, 88, 653–664. [Google Scholar] [CrossRef] [PubMed]
  87. Prather, R.S.; Lorson, M.; Ross, J.W.; Whyte, J.J.; Walters, E. Genetically Engineered Pig Models for Human Diseases. Annu. Rev. Anim. Biosci. 2013, 1, 203–219. [Google Scholar] [CrossRef]
  88. Wolf, E.; Braun-Reichhart, C.; Streckel, E.; Renner, S. Genetically engineered pig models for diabetes research. Transgenic Res. 2014, 23, 27–38. [Google Scholar] [CrossRef] [PubMed]
  89. Kyrova, K.; Stepanova, H.; Rychlik, I.; Polansky, O.; Leva, L.; Sekelova, Z.; Faldyna, M.; Volf, J. The response of porcine monocyte derived macrophages and dendritic cells to SalmonellaTyphimurium and lipopolysaccharide. BMC Vet. Res. 2014, 10, 244. [Google Scholar] [CrossRef] [PubMed]
  90. Pomorska-Mól, M.; Czyżewska-Dors, E.; Kwit, K.; Pejsak, Z. Enrofloxacin in therapeutic doses alters cytokine production by porcine PBMCs induced by lipopolysaccharide. Drug Chem. Toxicol. 2017, 40, 295–299. [Google Scholar] [CrossRef]
  91. Adler, M.; Murani, E.; Brunner, R.; Ponsuksili, S.; Wimmers, K. Transcriptomic Response of Porcine PBMCs to Vaccination with Tetanus Toxoid as a Model Antigen. PLoS ONE 2013, 8, e58306. [Google Scholar] [CrossRef]
  92. Blome, S.; Gabriel, C.; Beer, M. Pathogenesis of African swine fever in domestic pigs and European wild boar. Virus. Res. 2013, 173, 122–130. [Google Scholar] [CrossRef]
  93. Canter, J.A.; Aponte, T.; Ramirez-Medina, E.; Pruitt, S.; Gladue, D.P.; Borca, M.V.; Zhu, J.J. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022, 14, 1249. [Google Scholar] [CrossRef]
  94. Costers, S.; Lefebvre, D.J.; Goddeeris, B.; Delputte, P.L.; Nauwynck, H.J. Functional impairment of PRRSV-specific peripheral CD3 + CD8 high cells. Vet. Res. 2009, 40, 46. [Google Scholar] [CrossRef]
  95. Franzoni, G.; Pedrera, M.; Sánchez-Cordón, P.J. African Swine Fever Virus Infection and Cytokine Response In Vivo: An Update. Viruses 2023, 15, 233. [Google Scholar] [CrossRef]
  96. Islam, M.d.A.; Große-Brinkhaus, C.; Pröll, M.J.; Uddin, M.J.; Aqter Rony, S.; Tesfaye, D.; Tholen, E.; Hoelker, M.; Schellander, K.; Neuhoff, C. PBMC transcriptome profiles identifies potential candidate genes and functional networks controlling the innate and the adaptive immune response to PRRSV vaccine in Pietrain pig. PLoS ONE 2017, 12, e0171828. [Google Scholar] [CrossRef] [PubMed]
  97. Yang, T.; Zhang, F.; Zhai, L.; He, W.; Tan, Z.; Sun, Y.; Wang, Y.; Liu, L.; Ning, C.; Zhou, W.; et al. Transcriptome of Porcine PBMCs over Two Generations Reveals Key Genes and Pathways Associated with Variable Antibody Responses post PRRSV Vaccination. Sci. Rep. 2018, 8, 2460. [Google Scholar] [CrossRef] [PubMed]
  98. Reiske, L.; Schmucker, S.; Steuber, J.; Stefanski, V. Glucocorticoids and Catecholamines Affect In Vitro Functionality of Porcine Blood Immune Cells. Animals 2019, 9, 545. [Google Scholar] [CrossRef]
  99. Li, Y.; Díaz, I.; Martín-Valls, G.; Beyersdorf, N.; Mateu, E. Systemic CD4 cytotoxic T cells improve protection against PRRSV-1 transplacental infection. Front. Immunol. 2023, 13, 1020227. [Google Scholar] [CrossRef] [PubMed]
  100. Olesen, A.S.; Kodama, M.; Skovgaard, K.; Møbjerg, A.; Lohse, L.; Limborg, M.T.; Bøtner, A.; Belsham, G.J. Influence of African Swine Fever Virus on Host Gene Transcription within Peripheral Blood Mononuclear Cells from Infected Pigs. Viruses 2022, 14, 2147. [Google Scholar] [CrossRef]
  101. Fort, M.; Fernandes, L.T.; Nofrarias, M.; Díaz, I.; Sibila, M.; Pujols, J.; Mateu, E.; Segalés, J. Development of cell-mediated immunity to porcine circovirus type 2 (PCV2) in caesarean-derived, colostrum-deprived piglets. Vet. Immunol. Immunopathol. 2009, 129, 101–107. [Google Scholar] [CrossRef]
  102. Knoetig, S.M.; Summerfield, A.; Spagnuolo-Weaver, M.; McCullough, K.C. Immunopathogenesis of classical swine fever: Role of monocytic cells. Immunology 1999, 97, 359–366. [Google Scholar] [CrossRef]
  103. Li, J.; Yu, Y.J.; Feng, L.; Cai, X.B.; Tang, H.B.; Sun, S.K.; Zhang, H.Y.; Liang, J.J.; Luo, T.R. Global transcriptional profiles in peripheral blood mononuclear cell during classical swine fever virus infection. Virus. Res. 2010, 148, 60–70. [Google Scholar] [CrossRef]
  104. Pensaert, M. Viremia and effect of fetal infection with porcine viruses with special reference to porcine circovirus 2 infection. Vet. Microbiol. 2004, 98, 175–183. [Google Scholar] [CrossRef]
  105. Hohnstein, F.S.; Meurer, M.; de Buhr, N.; von Köckritz-Blickwede, M.; Baums, C.G.; Alber, G.; Schütze, N. Analysis of Porcine Pro- and Anti-Inflammatory Cytokine Induction by S. suis In Vivo and In Vitro. Pathogens 2020, 9, 40. [Google Scholar] [CrossRef]
  106. Wichgers Schreur, P.J.; Rebel, J.M.J.; Smits, M.A.; van Putten, J.P.M.; Smith, H.E. Lgt Processing Is an Essential Step in Streptococcus suis Lipoprotein Mediated Innate Immune Activation. PLoS ONE 2011, 6, e22299. [Google Scholar] [CrossRef]
  107. Hara, H.; Long, C.; Lin, Y.J.; Tai, H.C.; Ezzelarab, M.; Ayares, D.; Cooper, D.K.C. In vitro investigation of pig cells for resistance to human antibody-mediated rejection. Transpl. Int. 2008, 21, 1163–1174. [Google Scholar] [CrossRef] [PubMed]
  108. Petersen, B.; Ramackers, W.; Tiede, A.; Lucas-Hahn, A.; Herrmann, D.; Barg-Kues, B.; Schuettler, W.; Friedrich, L.; Schwinzer, R.; Winkler, M.; et al. Pigs transgenic for human thrombomodulin have elevated production of activated protein C. Xenotransplantation 2009, 16, 486–495. [Google Scholar] [CrossRef] [PubMed]
  109. Phelps, C.J.; Koike, C.; Vaught, T.D.; Boone, J.; Wells, K.D.; Chen, S.H.; Ball, S.; Specht, S.M.; Polejaeva, I.A.; Monahan, J.A.; et al. Production of α1,3-Galactosyltransferase-Deficient Pigs. Science 2003, 299, 411–414. [Google Scholar] [CrossRef]
  110. Butler, J.R.; Martens, G.R.; Estrada, J.L.; Reyes, L.M.; Ladowski, J.M.; Galli, C.; Perota, A.; Cunningham, C.M.; Tector, M.; Tector, A.J. Silencing porcine genes significantly reduces human-anti-pig cytotoxicity profiles: An alternative to direct complement regulation. Transgenic Res. 2016, 25, 751–759. [Google Scholar] [CrossRef] [PubMed]
  111. Cooper, D.K.C.; Dorling, A.; Pierson, R.N.; Rees, M.; Seebach, J.; Yazer, M.; Ohdan, H.; Awwad, M.; Ayares, D. α1,3-Galactosyltransferase Gene-Knockout Pigs for Xenotransplantation: Where Do We Go From Here? Transplantation 2007, 84, 1–7. [Google Scholar] [CrossRef]
  112. Estrada, J.L.; Martens, G.; Li, P.; Adams, A.; Newell, K.A.; Ford, M.L.; Butler, J.R.; Sidner, R.; Tector, M.; Tector, J. Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/β4GalNT2 genes. Xenotransplantation 2015, 22, 194–202. [Google Scholar] [CrossRef]
  113. Gock, H.; Nottle, M.; Lew, A.M.; d’Apice, A.J.F.; Cowan, P. Genetic modification of pigs for solid organ xenotransplantation. Transplant. Rev. 2011, 25, 9–20. [Google Scholar] [CrossRef]
  114. Lai, L.; Kolber-Simonds, D.; Park, K.W.; Cheong, H.T.; Greenstein, J.L.; Im, G.S.; Samuel, M.; Bonk, A.; Rieke, A.; Day, B.N.; et al. Production of α-1,3-Galactosyltransferase Knockout Pigs by Nuclear Transfer Cloning. Science 2002, 295, 1089–1092. [Google Scholar] [CrossRef]
  115. Sachs, D.H.; Galli, C. Genetic manipulation in pigs. Curr. Opin. Organ Transplant. 2009, 14, 148–153. [Google Scholar] [CrossRef]
  116. Yamada, K.; Yazawa, K.; Shimizu, A.; Iwanaga, T.; Hisashi, Y.; Nuhn, M.; O’Malley, P.; Nobori, S.; Vagefi, P.A.; Patience, C.; et al. Marked prolongation of porcine renal xenograft survival in baboons through the use of α1,3-galactosyltransferase gene-knockout donors and the cotransplantation of vascularized thymic tissue. Nat. Med. 2005, 11, 32–34. [Google Scholar] [CrossRef] [PubMed]
  117. Paillot, R.; Laval, F.; Audonnet, J.-C.; Andreoni, C.; Juillard, V. Functional and phenotypic characterization of distinct porcine dendritic cells derived from peripheral blood monocytes. Immunology 2001, 102, 396–404. [Google Scholar] [CrossRef]
  118. Buermann, A.; Petkov, S.; Petersen, B.; Hein, R.; Lucas-Hahn, A.; Baars, W.; Brinkmann, A.; Niemann, H.; Schwinzer, R. Pigs expressing the human inhibitory ligand PD-L1 (CD274) provide a new source of xenogeneic cells and tissues with low immunogenic properties. Xenotransplantation 2018, 25, e12387. [Google Scholar] [CrossRef] [PubMed]
  119. Golbus, A.L.; Ochoa, B.V.; Hardy, W.A.; Helke, K.L.; Kavarana, M.N.; Kwon, J.H.; Rajab, T.K. Immunosuppressive regimens in porcine transplantation models. Transplant. Rev. 2022, 36, 100725. [Google Scholar] [CrossRef] [PubMed]
  120. Vaughan, A.N.; Malde, P.; Rogers, N.J.; Jackson, I.M.; Lechler, R.I.; Dorling, A. Porcine CTLA4-Ig Lacks a MYPPPY Motif, Binds Inefficiently to Human B7 and Specifically Suppresses Human CD4+ T Cell Responses Costimulated by Pig But Not Human B7. J. Immunol. 2000, 165, 3175–3181. [Google Scholar] [CrossRef]
  121. Hara, H.; Witt, W.; Crossley, T.; Long, C.; Isse, K.; Fan, L.; Phelps, C.J.; Ayares, D.; Cooper, D.K.C.; Dai, Y.; et al. Human dominant-negative class II transactivator transgenic pigs—Effect on the human anti-pig T-cell immune response and immune status. Immunology 2013, 140, 39–46. [Google Scholar] [CrossRef]
Figure 1. Protocol of PBMC isolation from peripheral blood of swine. Abbreviations: PBMC = peripheral blood mononuclear cells; PBS = phosphate-buffered saline; RBC = red blood cells; RPMI 1640 = Roswell Park Memorial Institute 1640 medium.
Figure 1. Protocol of PBMC isolation from peripheral blood of swine. Abbreviations: PBMC = peripheral blood mononuclear cells; PBS = phosphate-buffered saline; RBC = red blood cells; RPMI 1640 = Roswell Park Memorial Institute 1640 medium.
Jcm 14 03432 g001
Figure 2. Comparison of blood sample separation using 50 mL tube and SepMate™-50 tube. Abbreviations: B = B lymphocyte; Bas = basophils; DC = dendritic cells; Eos = eosinophils; Mo = monocytes; NK = natural killer cells; Neu = neutrophils; PBMC = peripheral blood mononuclear cells; RBC = red blood cells; T = T lymphocyte.
Figure 2. Comparison of blood sample separation using 50 mL tube and SepMate™-50 tube. Abbreviations: B = B lymphocyte; Bas = basophils; DC = dendritic cells; Eos = eosinophils; Mo = monocytes; NK = natural killer cells; Neu = neutrophils; PBMC = peripheral blood mononuclear cells; RBC = red blood cells; T = T lymphocyte.
Jcm 14 03432 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pietrzak, M.; Chaszczewska-Markowska, M.; Zemelka-Wiacek, M. Porcine Peripheral Blood Mononuclear Cells (PBMCs): Methods of Isolation, Cryopreservation, and Translational Applications in Human Studies. J. Clin. Med. 2025, 14, 3432. https://doi.org/10.3390/jcm14103432

AMA Style

Pietrzak M, Chaszczewska-Markowska M, Zemelka-Wiacek M. Porcine Peripheral Blood Mononuclear Cells (PBMCs): Methods of Isolation, Cryopreservation, and Translational Applications in Human Studies. Journal of Clinical Medicine. 2025; 14(10):3432. https://doi.org/10.3390/jcm14103432

Chicago/Turabian Style

Pietrzak, Magdalena, Monika Chaszczewska-Markowska, and Magdalena Zemelka-Wiacek. 2025. "Porcine Peripheral Blood Mononuclear Cells (PBMCs): Methods of Isolation, Cryopreservation, and Translational Applications in Human Studies" Journal of Clinical Medicine 14, no. 10: 3432. https://doi.org/10.3390/jcm14103432

APA Style

Pietrzak, M., Chaszczewska-Markowska, M., & Zemelka-Wiacek, M. (2025). Porcine Peripheral Blood Mononuclear Cells (PBMCs): Methods of Isolation, Cryopreservation, and Translational Applications in Human Studies. Journal of Clinical Medicine, 14(10), 3432. https://doi.org/10.3390/jcm14103432

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop