Abstract
Ex vivo organ perfusion (EVOP) is used for whole organ preservation, and the main focus is to improve the outcome of donor organs for transplantation. Recently, EVOP has found application in disease modeling, drug development, and tissue regeneration. We discuss progress in EVOP research involving small animal organs using benchtop and incubator-based EVOP systems, highlighting innovative designs of EVOP systems, technical specifications of each system, and their versatile applications across a range of research fields.
1. Introduction
Ex vivo organ perfusion (EVOP) systems aim to preserve whole organs outside of the body by maintaining them in a temperature-controlled environment and perfusing a solution through the vasculature. Over the past six decades, ex vivo whole-organ perfusion has undergone significant evolution from early exploration in research labs to its current clinical use for organ preservation prior to transplantation. One of the first EVOP systems was developed by Carrel and Lindbergh in 1935 to study the nutritional needs of organs including the thyroid, suprarenal gland, ovary, spleen, heart, and kidney [1]. It incorporated constant perfusion with a range of perfusion solutions, sterile conditions, and constant temperature to achieve optimal organ health [1]. Carrel and Lindbergh's work laid the foundation for future ex vivo perfusion research which further progressed in the 1960s, in parallel with advancements in surgical techniques used in organ transplantation.
Today, EVOP is used in transplant organ preservation [2, 3], expanding the pool of transplantable organs and reducing the donor organ shortage. For decades, EVOP research has relied mainly on large animal models for improving organ transplantation [4]. Compared to rodent models, porcine models have a greater similarity to humans in their genome sequence and immune system which are imperative when studying allogenic transplantation [4]. Their large size also facilitates easier cannulation of blood vessels and execution of transplant procedures [5]. While porcine models serve as ideal platforms for studying organ transplantation, their use in other applications—such as disease modeling, drug development, and tissue repair—remain constrained, largely due to high costs and limited availability. Small animal organs are more accessible, more cost-effective, and amenable to high-throughput modeling [5]. The widespread availability of genetically engineered models in rodents, especially in mice, also makes them better suited for disease modeling, compared to large animals. Therefore, small animal organs are valuable models for studying diseases, evaluating therapeutics, and overall advancing scientific pursuits.
2. EVOP in Small Animal Models
Scientific investigation has relied on diverse in vitro, in vivo, and ex vivo platforms, each offering unique benefits. In vitro cellular models are user-friendly, scalable for high-throughput assays [6], and can serve as patient-specific models [7]. Nevertheless, they fail to fully replicate organ physiology due to the absence of cellular heterogeneity and proper structural organization [8]. Conversely, in vivo animal models can represent human physiology; however, the complexity of these models poses challenges such as real-time visualization and assessment which require invasive procedures [5]. To address some of these challenges, scientists have developed three-dimensional multicellular in vitro–based models such as organoids and organ-on-chip devices to introduce more complexity to conventional cell cultures while maintaining controlled conditions for easier manipulation [9]. While organoids represent a more heterogeneous cell population, they do not contain all tissue layers and cell types [10]. Most organoid models also represent a fetal stage, making them less ideal for studying adult diseases [5]. Organ-on-chip devices also face challenges as most have been designed without taking into account the appropriate size and flow rate of each organ or full cell composition [11]. Other complex multicellular in vitro models such as tissue explants and organ slices contain the microscopic anatomical features specific to organs. However, their viability in culture is limited to shorter durations and the culture conditions do not accurately mimic physiological environments. EVOP models can address these shortcomings by mimicking physiologic conditions more accurately, while reducing the complexity of in vivo models [9]. These perfusion systems can isolate individual organs making it easier to study specific biological processes without systemic effects [5]. Compared to in vivo models, EVOP models are less costly and less time-consuming [5]. They also allow for real-time sampling of perfusion solutions for monitoring organ health [5]. Overall, EVOP systems represent isolated systems that closely recapitulate the cellular and the extracellular components of a whole organ. Owing to these benefits, EVOP platforms have gained much attention in a wide range of applications including disease modeling, cell therapy, drug testing, and tissue regeneration.
This review aims to explore the literature on EVOP systems designed for gut, lungs, kidneys, and pancreas, in small animals, focusing on applications excluding transplantation.
3. EVOP System Components
EVOP systems are designed to replicate physiologic conditions in order to sustain the viability of an organ ex vivo. To achieve dynamic flow, EVOP systems are often equipped with an inlet for antegrade perfusion of the organ, as well as an outlet for drainage of the perfusate solution. A peristaltic pump is usually used to achieve this dynamic flow. Perfusion systems can be designed to adjust for varying physiological measurements such as (i) oxygenation, (ii) flow dynamics, and (iii) temperature. Oxygenation in EVOP systems is important to maintain [12]. In addition to tubing material, the diameter of the tubing is important to the flow rate of the perfused organ. Flow rate contributes to the nutrient and oxygen supply of the tissue. Optimal flow rate is crucial to help meet the demands of the perfused tissue. To control for the temperature, some EVOP systems are kept in a temperature-controlled incubator. For benchtop EVOP systems, temperature can be manipulated by hot plates or circulating water around the tissue and perfusion media chambers.
4. Perfusion Solutions
There are several types of perfusion solutions that are commonly used in EVOP in both small and large animals. Perfusion solutions mimic physiological conditions necessary for organ function. These solutions typically contain: (a) electrolytes to maintain cellular ionic balance, (b) nutrients such as vitamins and amino acids to support organ function, (c) buffers to maintain desired pH levels (in the absence of CO2), (d) antibiotics to prevent contamination, and (e) glucose to provide a source of energy, and may include other supplements such as oxygen carriers, knockout serum or fetal bovine serum (FBS) [13]. One of the most common perfusion solutions is the Krebs–Henseleit solution [14]. Krebs solution contains calcium chloride, potassium chloride, sodium chloride, as well as magnesium sulfate, glucose, and sodium bicarbonate buffer or HEPES [14, 15]. Generally, if the perfusion system is set up inside a tissue-culture incubator with a CO2 source, sodium bicarbonate and HEPES buffers are not required as a pH buffer. However, in the absence of CO2—for instance, in most benchtop EVOP systems—these buffers are required. STEEN, which is most commonly used for perfusion of lungs in large animals and human organs, has recently been applied to kidney perfusion as well [16]. STEEN solution contains human serum albumin which maintains osmotic pressure, as well as dextran 40 which inhibits leukocyte adhesion to the endothelium [17, 18]. Both serum albumin and dextran 40 also maintain optimal oncotic pressure to prevent edema [18]. Waymouth's media is another commonly used perfusate in preclinical EVOP. Waymouth's media is a serum-free medium that contains essential amino acids, vitamins, salts, and glucose and is commonly used in liver perfusion [19, 20]. In the context of kidneys, renal epithelial cell (REC) media has been used for ex vivo kidney perfusion in small animals [5]. REC contains several growth factors that enhance kidney viability and allow long-term perfusion in preclinical models. From a clinical perspective, Ringer's lactate—an isotonic salt solution—has been used for ex vivo perfusion of discarded human kidneys, demonstrating clinical feasibility. Ringer's solution lacks the necessary nutrients and growth factors, making it suitable for clinical EVOP with shorter perfusion times, but not for preclinical models that require long-term perfusion [21].
Oxygenation can be achieved by several means: carbogen gas, red blood cells (RBCs), hemoglobin-based oxygen carriers (HBOCs) which contain hemoglobin in a cell-free suspension, or whole blood perfusate [22, 23]. Although effective, the utility of RBCs is hampered by their limited availability, high costs, short shelf life, and risks of infection transmission and hemolysis [22]. Synthetic HBOCs, however, are more accessible, have reduced risks of infections, and provide greater oxygenation compared to RBCs [24]. Nevertheless, studies have shown that HBOCs can react to nitric oxide, leading to hypertension and vasoconstriction [24]. Further investigation is warranted to advance the clinical application of HBOCs. Alternatively, whole blood perfusate can be used due to its high concentration of RBCs. When compared to RBCs and HBOCs, whole blood perfusate was observed to be superior in tissue preservation [25]. However, whole blood perfusates contain leukocytes which can be implicated in ischemia-reperfusion (I/R) injury of the tissue [26]. Leukocyte filtering improves tissue function and reduces cell death.
Overall, advancement in EVOP systems happens in parallel to optimization, refinement, and customization of perfusion solutions. The primary goal of the perfusate is to enhance preservation and function of organs ex vivo in order to prolong the duration of experimental procedures.
5. Small Animal EVOP Systems
5.1. Lung
Numerous studies have applied EVOP for lung disease modeling purposes (Table 1). EVOP has been used extensively to model pulmonary I/R injury in small animals. Maxey et al. used an isolated perfused lung model to study the effects of tumor necrosis factor-alpha (TNFα) production by resident lung cells in pulmonary I/R [27]. Harvested lungs from wildtype and TNFα-deficient mice were cannulated at the inferior vena cava and trachea for ex vivo media perfusion and ventilation, respectively [27]. The organs were kept within a benchtop bioreactor by Hugo Sachs Elektronik-Harvard Apparatus for a total of 120 min [27]. To induce I/R injury, perfusion was arrested and ventilation was maintained at hypoxic conditions [27]. After 60 min of ischemia, the organ was reperfused with Krebs solution and ventilation was returned to room air. Using this platform, they analyzed lung weight, lung injury score, pulmonary arterial and venous pressures, and pulmonary compliance (the ability of lungs to expand) [27]. The authors found that, upon exposure to I/R, perfused lungs from TNFα-deficient mice showed significantly lower histologic injury, reduced pulmonary edema and pulmonary artery pressure, and improved pulmonary compliance compared to wildtype [27]. This indicates that TNF-α released from resident lung cells is a key initiating factor in lung I/R injury. This ex vivo perfusion model allowed for continuous real-time monitoring of arterial and venous pressures during organ reperfusion providing valuable insights on progression of I/R injury. The model also allows researchers to study the role of lung-derived TNF-α in isolation, hence avoiding confounding factors from systemic proinflammatory responses that might contribute to pulmonary I/R injury.
Table 1.
Summary table listing all publications discussed in this manuscript on ex vivo lung perfusion.
| Study | Animal species/strain | Perfusion solution | Tubing type/length | Use of oxygenators | Flow rate | Length of perfusion | Temperature (°C) | Application | Functional tests performed | Cannulation site |
|---|---|---|---|---|---|---|---|---|---|---|
| Lung | ||||||||||
| Benchtop | ||||||||||
| Maxey, 2004 | C57BL/6 mice (adult males) | Krebs–Henseleit buffer containing 2% albumin, 0.1% glucose, and 0.3% HEPES (335–340mOsm/kg) | Not described | No | 2 mL/min | 60 min | 37°C | Modeling pulmonary I/R injury | Lung weight, lung injury score, pulmonary artery and vein pressures, airway resistance, pulmonary compliance | Trachea for ventilation; inferior vena cava for media perfusion |
| Zhao, 2006 | C57BL/6 mice (adult males) | Krebs–Henseleit buffer containing 2% albumin, 0.1% glucose, and 0.3% HEPES (335–340mOsm/kg) | Not described | No | 60 μL/g body weight/min | 60 min | 37°C | Investigating the role of alveolar macrophages in pulmonary I/R injury | Lung weight, cytokine/chemokine expression (RT-PCR, ELISA), lung vascular permeability, neutrophil quantification, pulmonary artery pressure, pulmonary compliance | Trachea for ventilation; inferior vena cava for media perfusion |
| Francioli, 2017 | Sprague–Dawley rat (300 g) | Steen® solution (Xvivo perfusion, Göteborg, Sweden, pH 7.4) | Not described | Gas-exchange membrane | Not described | 4 h | 37°C | Testing of pharmaceutical drug pyrrolidine dithiocarbamate (PDTC) during warm ischemia | Physiological variables (PA pressure, arterial pressure, vascular resistance, compliance), protein concentration and lactate dehydrogenase activity in bronchoalveolar lavage, pro-inflammatory cytokine levels, NF-kB expression | Pulmonary artery for media perfusion; trachea for ventilation |
| Wang, 2016 | Sprague–Dawley rat (400 g) | Steen solution (Xvivo perfusion, Göteborg, Sweden, pH 7.4) | Not described | Gas-exchange membrane | Not described | 3 h | 37°C | Effect of MnTBAP and 3-AB on the lung during warm ischemic injury | Physiological variables (PA pressure, arterial pressure, vascular resistance, compliance), protein concentration and lactate dehydrogenase activity in bronchoalveolar lavage, pro-inflammatory cytokine levels, histology | Pulmonary artery for media perfusion; trachea for ventilation |
| Incubator | ||||||||||
| Mishra, 2012 | Sprague–Dawley rats (6-12-week-old male) | RPMI | Silicone (3 m) | No | 6 mL/min | 7–14 days | 37°C | Modeling human lung cancer on a decellularized rat lung matrix | Histology, DNA extraction, immunohistochemistry | Trachea for cell seeding, pulmonary artery for media perfusion |
| Ahmadipour, 2021 | C57BL/6 mice (12-14-week-old males) | High glucose content DMEM containing 10% FBS and 1% antibiotics | Not described | No | 1.5 mL/min (vascular perfusion rate); 40 breaths/min with a tidal volume of 300 μL (ventilation rate) | 3 days | 37°C | Describing a novel ventilation protocol for enhanced recellularization of rat lung scaffolds | Histology, immunofluorescence staining, immunohistochemistry, RT-PCR | Trachea for liquid ventilation and cell seeding, pulmonary artery for media perfusion |
| Engler, 2018 | Sprague–Dawley rat (200 g) | DMEM high glucose with 10% FBS, 100 U/mL penicillin, 100 μg/mL streptomycin, 3 μg/mL amphotericin B and 50 μg/mL gentamicin | Silicone (1.5 m) | Hollow fiber cartridge oxygenator | 4 mL/min (perfusion rate); 4, 6, 8, 16, or 32 mL/min (oxygenator flow rate) | 24 h | 37°C | Mathematical modeling and characterization of oxygen requirements of whole lung tissues | Histology, immunofluorescence staining, whole lung cell count, total lung weight, single cell oxygen consumption rate, glucose consumption, lactate production | Pulmonary artery for media perfusion |
Several incubator-based EVOP systems have been developed to study the lungs in small animal models. In particular, in 2021, Ahmadipour et al. designed an EVOP system that achieved both vascular perfusion and liquid ventilation of mouse lungs via cannulation of pulmonary artery and trachea, respectively (Figure 1) [28]. They created a novel negative-pressure ventilation system that allowed control over tidal volume and achieved a physiological ventilation rate [28]. Using this platform, they studied the effect of higher ventilation rate (40 breaths per minute) on recellularization of mouse lungs. Decellularized mouse lungs were placed inside the EVOP system in a supine orientation (mimicking in vivo positioning) [28]. Lungs were then seeded with human bronchial epithelial cells and cultured for 3 days [28]. They showed that at higher ventilation rates compared to lower ventilation rates, lungs had an increased cell surface coverage, higher cell numbers, as well as significantly higher number of proliferative cells [28]. This innovative EVOP design allows for precise control of tidal volume and shows the importance of physiologically relevant mechanical cues for recellularization of lung grafts. Similar to this group, other researchers have addressed other important challenges in designing EVOP systems that mimic physiologic environments. Strict oxygen control is vital: hypoxia triggers apoptosis, whereas hyperoxia causes damage or dedifferentiation. This study introduced a tunable whole-lung bioreactor that adjusts oxygen delivery in real time to match metabolic demand. A calibrated mathematical model, validated with rat lungs, accurately predicted gas exchange, maintaining stable dissolved-oxygen levels, thus offering noninvasive insight into tissue metabolism [29].
Figure 1.

Graphical representation of a lung EVOP system developed by Ahmadipour et al. [28]. The system consists of two reservoirs, the main reservoir where the lung is perfused and the ventilation reservoir. The main reservoir is filled completely with media. A syringe pump continuously pumps media into the main reservoir, with any extra media flowing into the ventilation reservoir. The trachea is perfused from the ventilation reservoir media, while the pulmonary artery is perfused via a peristaltic pump that circulates the main reservoir media. The EVOP system was maintained in an incubator at 37°C (95% air; 5% CO2).
Overall, there has been tremendous progress in ex vivo lung perfusion in both large animals, and more recently in small animal models. These models have been applied for studying various lung diseases including I/R injury and cancer, as well as for decellularization and tissue engineering purposes. Technical innovations have led to customizable EVOP platforms and data acquisition systems that support lung mechanics and provide high-level hemodynamic metrics. These modern systems offer real-time vascular pressure tracing, monitor respiratory rates, allow for easy assessment of gas exchange and injury biomarkers, and overall allow for highly controllable and reproducible perfusion circuits [30, 31]. Another state-of-the-art advancement has been the recent development of a low-volume ex situ lung perfusion (ESLP) system. This platform enables both single- and double-lung applications and requires only 17 mL of perfusate solution [32] within the circuit. This system not only reduces the amount and cost of perfusate significantly but also minimizes the number of animals needed. Although ex vivo lung perfusion research in small animals has made significant advancements, several critical gaps still remain. Currently, one of the major limitations in EVOP systems is the short perfusion times. Most described systems in rodent models can support up to 4–5 h of perfusion without damage to organ viability. In large animal models, this has been extended to 12 h. Extended perfusion times are needed for more accurate disease modeling purposes as well as therapeutic interventions. Another challenge of ex vivo lung perfusion in small animals is higher risk of atelectasis. Rodents have smaller airways, more delicate lung structures, and greater surface tension all of which make them more susceptible to atelectasis [33, 34]. To combat this, more precise ventilatory support is needed during the perfusion.
We believe that future work on ex vivo lung perfusion systems in small animals should be aimed at optimizing perfusate solutions and standardizing perfusion protocols. Currently, research on acellular and cellular-based perfusate solutions is more advanced in a clinical context compared to a preclinical small animal setting. In addition, there is a need to establish a consistent approach for ex vivo lung perfusion in small animals. This can ensure reproducibility and consistency within the field.
5.2. Kidneys
Ex vivo perfusion of kidneys has a long history spanning several decades (Table 2). In 1967, Nishiitsutsuji-Uwo et al. developed an isolated perfused rat kidney model based on previously published perfused models of rat liver [35, 36]. Using this system, they studied the metabolic activity of kidneys and found that under normal physiological levels, both glucose consumption and acetoacetate oxidation are important sources of fuel for basic respiration but not for secretion [35, 36]. They also discovered that glutamine production remained linear despite the decline of other additional products, such as aspartate, which is formed from the same precursor, glutamate [35, 36]. This demonstrated that the kidneys were an important source of the body's glutamine. The same perfusion apparatus was then adapted by Ross et al. in 1973 for their study of sodium reabsorption in an isolated rat kidney, specifically examining the preferred energy source for this process [37]. They optimized the perfusion media, enabling improved sodium reabsorption and glomerular filtration rate—markers of kidney functionality—that more closely resemble in vivo conditions [37]. It was found that glucose, not fatty acids, is the preferred energy source for sodium reabsorption [37]. Later in 2008, Rosenberger et al. adapted the same EVOP system to study acute kidney injury in diabetic rats [38]. Diabetic kidneys exhibited impaired function under hypoxic stress induced by the perfusion system, compared to the control [38]. This was evidenced by decreased glomerular filtration rate and sodium reabsorption, along with increased potassium secretion [38]. Additionally, there was extensive renal morphology damage, including severe tubular necrosis and collecting duct swelling [38]. Although functional impairment was observed in the perfusion system, it was not shown in vivo, as diabetic kidneys showed resilience to induced hypoxic stress. This phenomenon may be explained by a hypoxia-adaptation mechanism in the kidneys. The EVOP system allowed for this observation by inducing extreme hypoxic stress and preventing activation of this mechanism, which was difficult to achieve in vivo as it resulted in a high mortality.
Table 2.
Summary table listing all publications discussed in this manuscript on ex vivo kidney perfusion.
| Study | Animal species/strain | Perfusion solution | Tubing type/length | Use of oxygenators | Flow rate | Length of perfusion | Temperature (°C) | Application | Functional tests performed | Cannulation site |
|---|---|---|---|---|---|---|---|---|---|---|
| Kidney | ||||||||||
| Benchtop | ||||||||||
| Nishiitsutsuji-Uwo, 1967 | Wistar rats (adult males) | Krebs–Henseleit saline, 4.5% bovine serum albumin | Portex based | Yes, but details not specified | 16–30 mL/min | < 2 h (dependent on test) | 38°C–40°C | Studying metabolic activity of the kidney | Metabolism, oxygen consumption, glucose synthesis, creatinine clearance, urine analysis | (Vascular) renal artery and vein; ureter |
| Ross, 1973 | Wistar rats (adults) | Krebs–Henseleit high-bicarbonate saline, 10% bovine serum albumin (mixed with other solution dependent on test) | Not described | 95% O2; 5% CO2 | 34.5 mL/min | > 4 h | Not described | Studying energy sources for sodium reabsorption | Physiological variables (metabolism, oxygen consumption, glucose synthesis, creatinine clearance, urine analysis, glomerular filtration rate), sodium absorption | (Vascular) renal artery and vein; ureter |
| Rosenberger, 2008 | Sprague-Dawley rats (adult males) | Modified Krebs–Ringer–Henseleit solution, 6.7 g/dL bovine serum albumin, 20 amino acids | Not described | 95% O2; 5% CO2 | Not described | 90 min | 38°C | Studying acute kidney injury in diabetic rats | Physiological variables (creatinine clearance, plasma creatinine, urine analysis, sodium absorption, potassium excretion, glomerular filtration rate, necrosis), kidney morphology | (Vascular) renal artery and vein; ureter |
| Hori, 1993 | Wistar rats (adult males) | Modified Krebs–Henseleit bicarbonate buffer, bovine erythrocytes,5% bovine serum albumin,8 amino acids,5 nM glucose,3% mannitol, and 100 μg/mL creatinine | Not described | 95% O2; 5% CO2 | 5 mL/min | 15–30 min | Not described | Studying mechanism of digoxin excretion | Physiological variables (perfusate flow rate, urine flow rate, glomerular filtration rate, sodium and glucose fractional reabsorption), renal tubular secretion | (Vascular) renal artery and inferior vena cava; ureter |
| Yokota, 2018 | Wistar rats (12 week old males) | Modified Krebs–Henseleit solution ,6 g% bovine serum albumin, 8.3 mM urea, 1.25 mM inulin, and 5.55 mM glucose | Not described | Silastic membrane oxygenator | 20 mL/min | 90 min | 37°C | Studying the metabolic effects of high fructose intake | Renal perfusion pressure, renal vascular resistance, osmolar clearance, glomerular filtration rate, urinary flow | (Vascular) renal artery and vein; ureter |
| Mahboub, 2020 | Lewis rats (adult males) | Williams medium E containing 1000 U/L heparin, 15% albumin, 1000 µmol/L creatinine, and 25% HBOC-201 | Silicone based | 95% O2; 5% CO2 | ≥ 8 mL/min (to maintain physiological pressure) | 90 min (rewarming) then 120 min (reperfusion) | 10°C–37°C | Studying the effects of hemoglobin based oxygen carriers during gradual kidney rewarming | Physiological variables (ultrafiltrate production, glomerular filtration rate, sodium reabsorption, pH, lactate levels), renal artery resistance, weight, oxygen consumption, energy, kidney morphology | (Vascular) renal artery; ureter |
| Incubator | ||||||||||
| Uzarski, 2015 | Sprague-Dawley rats (adult males) | (Decellularization) Triton X-100, sodium dodecyl sulfate solution; (recellularization) DMEM/F-12, 10% FBS, 1% penicillin/streptomycin | Silicone based | 5% CO2 | 4 mL/min | 7 days | 37°C | Studying the effects of seeding canine or human derived renal cells onto decellularized rats kidney | Hydrostatic pressure, metabolic activity, kidney morphology, injury (kidney injury molecule 1) | (Vascular) renal artery; ureter |
| Won, 2022 | CD1 mice (adults) | (Media 1) Dulbecco's modified Eagle's medium, 10% fetal bovine serum; (media 2); renal epithelial cell media | Silicone based | 18.6% O2; 5% CO2 | 1.1 mL/min | 9 days | 37°C | Mathematically designed EVOP system for improving kidney longevity | Oxygen consumption, urine analysis, kidney morphology, immunostaining | (Vascular) renal artery; ureter |
An ex vivo kidney perfusion apparatus was designed and built by Won et al. in 2022 using mathematical modeling (Figure 2) [5]. Using a complementary error function and Henry's law, they predicted oxygen flux in the perfusion system and subsequently calculated the height of the organ chamber, the length of silicone tubing, and the flow rate required to maintain sufficient oxygenation of the kidney through atmospheric air [5]. The authors carefully developed a model that optimized surface area of the medium for sufficient oxygenation within the media reservoir [5]. Based on their calculations, they then fabricated an organ chamber containing a mouse kidney with the renal artery and the ureter both cannulated [5]. Kidneys were perfused with either DMEM +10% FBS or REC media for up to 9 days [5]. The experimentally measured oxygen levels of both arterial blood at the tissue level and at the vein exceeded the minimum physiological requirements, indicating that the mathematical modeling achieved the desired oxygen levels [5]. Urine analysis of total protein, albumin, and glucose levels relative to the concentrations found in the media indicated that perfused kidneys were properly functioning after 7 days. Low levels of total protein, albumin, and glucose were measured in the urine which implies that nutrients were absorbed, filtered by glomeruli, and reabsorbed by the proximal tubules [5]. This indicates accurate physiological processing of nutrients by the perfused kidneys. Furthermore, histology and immunofluorescence showed that kidneys remained healthy for up to 7 days ex vivo in the DMEM+10% FBS media and up to 9 days in the REC media [5].
Figure 2.

Graphical representation of an incubator kidney EVOP system developed by Won et al. [5]. The tissue perfusion chamber was connected to a closed perfusion loop. Media was pumped through the renal artery of mouse kidneys using a peristaltic pump. The renal vein drained directly into the media. An oxygen sensor was incorporated into the perfusion loop before the renal artery cannulation. The ureter was cannulated, and urine was collected via a syringe on day 7. The EVOP system was inside an incubator that maintained the temperature and oxygenation of the media.
Overall, kidney EVOP systems have undergone significant technical development. Small animal kidney EVOP systems have increased usage beyond organ transplantation. They are being utilized to investigate metabolic activity and nutrient source usage [35–37]. Furthermore, drug interactions [39] and hypoxic stress responses [38], difficult to study in vivo, have been observed using kidney EVOP systems. In comparison to other organ EVOP system's abilities to maintain tissue viability, small animal kidneys can be preserved for 7–9 days [5]. This ability expands the potential of disease modeling and mechanism investigations using kidney EVOP systems. However, this ability also highlights the technical aspects of EVOP systems important to optimizing the environment for the kidney. For example, continuous monitoring of perfusate flow rate in order to optimize oxygenation and meet the kidney's metabolic demand [5]. For future studies using kidney EVOP systems, adjustments to not only the perfusion media but also the monitoring systems may need to be made to ensure sufficient nutrient and oxygen supply to the tissue.
5.3. Gut
Gut ex vivo perfusion has been extensively documented (Table 3). In 2010, Lautenschlager et al. developed a perfused rat small bowel model to study the role of proinflammatory mediator platelet-activating factor (PAF) in intestinal edema [40]. Using a custom-made perfusion chamber from Hugo Sachs Elektronik-Harvard Apparatus, they assembled both vascular and luminal perfusion of the small bowel [40]. They demonstrated well-preserved gut viability after 4 h of perfusion by observing important functional parameters such as oxygen consumption, CO2 partial pressure, metabolism (lactate-to-pyruvate ratio), pH, cellular necrosis, and galactose uptake [40]. Additionally, intestinal morphology was preserved [40]. Administration of PAF led to vasoconstriction, inhibition of galactose uptake, as well as increased epithelial and endothelial permeability, which were reversed to normal levels after 30 min of perfusion [40]. Further, luminal effluent volume increased while the opposite was observed for vascular effluent volume. These findings emphasize the tightly controlled mechanisms of PAF in the intestine and the importance of luminal clearance as a defense mechanism. They also looked at underlying mechanisms of PAF-induced vasoconstriction and hyperpermeability and their involvement in intestinal failure [41].
Table 3.
Summary table listing all publications discussed in this manuscript on ex vivo gut perfusion.
| Study | Animal species/strain | Perfusion solution | Tubing type/length | Use of oxygenators | Flow rate | Length of perfusion | Temperature (°C) | Application | Functional tests performed | Cannulation site |
|---|---|---|---|---|---|---|---|---|---|---|
| Gut | ||||||||||
| Benchtop | ||||||||||
| Lautenschlager, 2010 | Wistar rats (adult females) | (Vascular) modified Krebs–Henseleit solution; (luminal) 114 mM NaCl, 5 mM KCl, 26 mM NaHCO3, 30 mM lactose, 5.55 mM glucose, 10 mM mannitol, and 0.8 mM glutamine (310–320 mOsm/L) | Tygon based | 95% O2; 5% CO2 | (Vascular) 7.5 mL/min; (luminal) 0.15 mL/min | 240 min | 36°C | Studying the role of platelet-activating factor in intestinal edema | Physiological variables (oxygen consumption, CO2 partial pressure, metabolism, pH, cellular necrosis, galactose uptake, mucosal surface scoring), luminal and vessel fluid measurements, mesenteric artery and vein pressure, luminal pressure, intestinal motility | (Vascular) superior mesenteric artery and hepatic portal vein; (luminal) proximal and distal small intestine |
| Boyle, 2016 | Wistar rats (adult females) | (Vascular) modified Krebs–Henseleit solution; (luminal) 114 mM NaCl, 5 mM KCl, 26 mM NaHCO3, 30 mM lactose, 5.55 mM glucose, 10 mM mannitol, and 0.8 mM glutamine (310–330 mOsm/L) | Tygon based | 95% O2; 5% CO2 | (Vascular) 7.5 mL/min; (luminal) 0.05 mL/min | 240 min | 37°C | Modeling Salmonella enteritis | Intestinal epithelial scoring, epithelial tight junction staining, pro-inflammatory cytokine levels, arterial pressure, intestinal absorption (galactose uptake), intestinal lumen fluid analysis | (Vascular) superior mesenteric artery and hepatic portal vein; (luminal) proximal and distal small intestine |
| Gagliardi, 2021 | Balb/c mice (8 weeks) | IMDM containing 20% Knockout serum replacement, 2% B-27,1% of N-2,1% L-glutamine, 1% non-essential amino acids, 1% HEPES | Not described | 95% O2; 5% CO2 | 99 μL/hr | 16 h | 37°C | Modeling celiac disease | MTT assay, unfolded protein response levels, gliadin derived peptides, intestinal permeability, pro-inflammatory cytokine levels, intestinal morphology | Proximal and distal small intestine |
| Gagliardi, 2021 | Balb/c mice (13 days) | IMDM containing 20% Knockout serum replacement, 2% B-27,1% of N-2,1% L-glutamine, 1% non-essential amino acids, 1% HEPES | Not described | 95% O2; 5% CO2 | 99 μL/h | 5 h | 37°C | Modeling inflammatory bowel disease | MTT assay, unfolded protein response levels, intestinal permeability, pro-inflammatory cytokine levels, intestinal morphology | Proximal and distal colon |
| Tan, 2020 | C57BL/6 mice (8–10 week old females) | (Extraluminal) modified Krebs solution; (intraluminal) Phosphate buffer solution | Not described | 95% O2; 5% CO2 | 30 μL/min | > 30 min (dependent on test) | 35°C | Studying the effects of distention and fatty acids on intestinal motility | Colonic motor complex activity, diameter of colon, intraluminal pressure and pressure amplitude, pellet expulsion monitoring | Proximal and distal colon |
| Schreiber, 2014 | Sprague-Dawley rats (adult) | (Vascular) Tyrode solution; (luminal) Tyrode solution | Not described | 95% O2; 5% CO2 | (Vascular) 3 mL/min; (luminal) 80 mL/h | > 30 min (dependent on test) | 37°C | Pharmacological testing and effects on intestinal motility | Jejunum intestinal motility, intestinal morphology | (Vascular) superior mesenteric artery and vein; (luminal) proximal and distal jejunal region |
| Incubator | ||||||||||
| Motherwell, 2019 | Wistar rats (adult males) | Culture media, 10% fetal bovine serum | Not described | 5% CO2 | 0.12 mL/min | 48 h | 37°C | Studying the effects of flow dynamics on angiogenesis | Vascular analysis (immunofluorescent imaging, diameter, velocity, shear stress, sprouting) | Main feeding artery and vein |
| Willi, 2022 | NG2DsRedBAC mice (6–12 months); C57BL/6 mice (6–12 months) | Minimum essential media, 1,5, or 10% fetal bovine serum, 1% penicillin-streptomycin | Silicone based | 5% CO2 | 1–1.5 mL/min | 48 h | 37°C | Studying viability and dynamics of intestinal vasculature | Vascular analysis (immunofluorescent imaging, density tortuosity), cell viability | Feeder artery |
In 2017, Yissachar et al. employed a silicone-based microfluidic chip that included six segments of mouse small intestine or colon (Figure 3) [42]. They utilized this perfusion system to evaluate intestinal immune and enteric nervous system (ENS) responses to bacteria. Tissue viability of the small intestine and colon was assessed through the analysis of morphology, mucus production, epithelial barrier integrity, proliferation, and enteric neurons [42].
Figure 3.

Graphical representation of a benchtop EVOP system developed by Yissachar et al. [42]. The tissue perfusion chamber consisted of six individual chambers for the cannulation of six intestines. Each intestinal chamber was connected to its own respective open perfusion loop. Media was perfused through the lumen and outside area of each individual intestine by synchronized intra- and extraluminal syringe pump inputs, respectively. The media was then withdrawn from synchronized intra- and extraluminal syringe pump outputs. The EVOP system was sealed and gassed using an oxygenator. Additionally, temperature was maintained using a heating plate.
Furthermore, they examined the immunocyte population present in the perfused tissues [42]. The viability of small intestine and colon was maintained for 2 and 24 h, respectively [42]. They then perfused germ-free immunocyte-activating bacteria intraluminally [42]. The perfusion system allowed them to observe immediate immune transcriptional changes in the small intestine and colon in response to bacteria. The ability to observe short-term transcriptional changes is not possible without the perfusion system, as the bacteria take days to fully develop within the gut in vivo. Additionally, they demonstrated that bacteria impact the ENS which subsequently affects immunocyte activation.
In addition to disease modeling, EVOP systems can be used to evaluate intestinal motility. Tan et al. evaluated the relationship between colonic distention (intraluminal pressure on intestinal tissue) and fatty acids (butyrate and propionate) and their role in induced intestinal motility in the isolated mouse colon [43]. They found that propionate reduced intestinal motility, but the effects were reversed upon increasing distention by adjusting the intraluminal pressure [43]. Intestinal motility increased proportional to distention. The same results were observed when butyrate was added after propionate, leading to enhanced intestinal motility [43]. However, this phenomenon of butyrate was observed at baseline distension. The effects of butyrate on intestinal motility were abolished in the presence of greater distension [43]. These findings highlight the importance of butyrate in cases of low intestinal motility that result from a fiber-deficient diet.
Several other groups have developed more compact ex vivo perfusion systems, complex enough to sustain organ function yet small enough to fit inside a standard tissue-culture incubator. Motherwell et al. designed a simple ex vivo model of rat intestinal mesenteric vasculature to study the effects of flow dynamics on angiogenesis [44]. After 48 h, static controls (no perfusion) had greater microvascular density compared to the perfused group. Additionally, invasive sprouting (into avascular regions) was greater in the perfused group while introverting (sprouting into central vascular regions) was greater in the static group [44]. These findings suggest that flow rate and shear stress greatly impact angiogenesis.
Similar to this model, Willi et al. developed a model of mouse intestinal mesentery by cannulating and perfusing a large mesenteric artery for 48 h [45]. They maintained viability after 48 h in 10% FBS-supplemented media but not in 1% or 5% FBS [45]. Despite the preserved tissue viability, there was significant reduction in microvasculature at 24 h and more so at 48 h [45]. Further analysis into this phenomenon revealed that microvasculature tortuosity remained high and constant over 48 h. This observation explains the preserved tissue viability, as tortuosity is a protection mechanism against increased intravascular pressure that could result in vascular damage.
In summary, intestinal EVOP systems are a useful platform that have been used to investigate molecular pathways41 and cellular interactions [42], as well as vasculature [44, 45] and motility dynamics [43] that are difficult to observe in vivo. Furthermore, they have been utilized to model diseases in a cost- and time-effective manner [46–48]. Despite the advancements of intestinal EVOPs, there are still limitations to their abilities due to the complex nature of the intestine. This complex nature involves interactions between the lumen, vasculature, ENS and muscular layer. To maintain these relationships ex vivo, bi-cannulation of both the vasculature and the lumen is important. Furthermore, the lumen contains bacteria and fungi that are essential to intestinal function in vivo. However, these microorganisms pose significant challenges to tissue viability ex vivo. To combat this, germ-free mice can be used but the microbiome–intestine crosstalk is lost. In addition to the overall complexity of the intestine, the small intestine and colon differ in both function and anatomy. This may add to the technical difficulties of maintaining intestinal tissue viability. For instance, one study was able to preserve the colon for 24 h, but only 2 h for the small intestine [42]. For future studies, the technical aspects of the EVOP system should be considered carefully for each component of the intestine. This includes the perfusion solution and flow dynamics that are respectively optimal for the small intestine, colon, and vasculature.
5.4. Pancreas
Ex vivo pancreas perfusion was first performed by Babkin and Starling in 1926 in a canine model using hypothermic machine perfusion [49, 50]. Many pancreas ex vivo perfusion studies followed (Table 4). Similar to other organs, early studies were not successful as they discovered several challenges during perfusion [51]. One major challenge is the fragile nature of the pancreas as it is a softer tissue organ and is composed mainly of exocrine glands that secrete digestive enzymes [52]. As a result, the pancreas is extremely sensitive to manipulation, and when injured, can secrete high levels of digestive enzymes (including protease and lipases) from its glands, leading to self-degradation of the tissue. Additional challenges seen in pancreas reperfusion include edema, hemorrhage, and venous congestion.
Table 4.
Summary table listing all publications discussed in this manuscript on ex vivo pancreas perfusion.
| Study | Animal species/strain | Perfusion solution | Tubing type/length | Use of oxygenators | Flow rate | Length of perfusion | Temperature (°C) | Application | Functional tests performed | Cannulation site |
|---|---|---|---|---|---|---|---|---|---|---|
| Pancreas | ||||||||||
| Benchtop/incubator | ||||||||||
| Single-pass method (Pegg, 1982) | WAG rats | Gelatin polypeptide Haemaccel + 5.4 mM calcium + glucose (5 mM for first hour, 30 mM for second hour) | Not described | Bubbled oxygen (95% O2, 5% CO2) | 2 mL/min | 2 h | 38°C | To test assays for measuring glucose-responsiveness | Insulin secretion assay (5 mM vs. 30 mM glucose) | Input: Aorta (below superior mesenteric artery) |
| Recirculation method (Pegg, 1982) | WAG rats | Gelatin polypeptide Haemaccel + 5.4 mM calcium + 5 mM glucose | Not described | Bubbled oxygen (95% O2, 5% CO2) | 60 mmHg (pressure) | 2 h | 38°C | To test assays for measuring glucose-responsiveness | Insulin secretion assay (5 mM vs. 30 mM glucose) | Input: Aorta (below superior mesenteric artery) |
| Loubatieres-Mariani, 1980 | Wistar rats | Krebs–Ringer bicarbonate buffer + 2 g/L purified bovine albumin + 0–3 g/L glucose | Not described | Bubbled oxygen (93% O2, 7% CO2) | 2.4 mL/min | 90 min | 28°C & 37.5°C | To test effect of temperature (hypothermia) on glucose responsiveness | Insulin secretion assay (following stimulation with high glucose, tolbutamide, acetylcholine, and glucagon) | N/A |
| Kojima, 1984 | Wistar rats | Na+; 131 mEq/1, lactate-: 28 mEq/L, K+: 4 mEq/1, Ca∗: 3 mEq/L, CI-: 110 mEq/1, Dextran 40:30.0 gr/1, glucose: 1 gr/1, pH: 8.2 + 0.2. T | Not described | Artificial lung | 10 mL/min | 6 h | 4°C (hypothermic) | To test the effect of hypothermia, hyperbaria, and oxygenation during pancreas preservation on islet health and function | Insulin secretion assay; transplantation into diabetic mice for reversal of diabetes | Input: celiac axis |
| Klempanauer, 1982 | Wistar rats | Protide gel solution, fructose-bicarbonate solution, modified cryoprecipitated plasma, Collins' C3 solution, Sack's solution | Not described | No | Not described | 24 h | 0°C–4°C | To demonstrate that a simpler approach, using initial perfusion with a chilled solution, following by storage at 0–4C is superior to other methods for 24 h perfusion | N/A | Input: Aorta (below superior mesenteric artery) |
| Mantke, 2009 | Wistar rats | Modified Krebs–Ringer bicarbonate | Not described | 95% O2, 5% CO2 | 1 mL/min (220 mmHg) | 80 min (60 min perfusion time, 20 min equilibration time) | 37°C (using heat exchanger) | To examine early mechanisms leading to acinar cell injury in acute pancreatitis | Amylase, lipase, and lactate dehydrogenase activity | Causal aorta (input), portal vein (output). Both cannulated with Teflon tube |
Various cold storage perfusates have been developed for the pancreas, with the most popular being the University of Wisconsin (UW) solution, which has become the gold standard preservation solution for the pancreas for 20 years [53]. The UW solution was created based on studies using the canine pancreas, where Wahlberg et al. demonstrated that this perfusate allowed for optimal control of edema allowing for preservation up to 24 h [54].
The first use of rodent pancreas in EVOP was performed by Loubatieres' group in 1980, using a Krebs–Ringer bicarbonate buffer including albumin and glucose, as well as bubbled oxygen and carbon dioxide [55]. They studied the effects of hypothermia on endocrine function of the pancreas by culturing the organ at normothermic (37.5°C) and hypothermic (28°C) temperatures. Insulin secretion was measured following stimulation by glucose, tolbutamide, and acetylcholine [55]. They found that under hypothermic temperatures, glucose-responsive insulin secretion was reduced, suggesting that normothermic perfusion is superior for maintaining normal endocrine function [55]. Shortly after, Pegg et al. generated a rat pancreas normothermic perfusion system named the “single-pass method” [56]. This system contained two separate circuits with different glucose concentrations, allowing for a switch between them without recirculation of the perfusate in order to easily assess insulin release upon changes in glucose concentration [56]. Pegg et al. demonstrated that the pancreas could be cultured for up to 2 h using their system with normal glycemic responsiveness and low levels of edema, as measured by wet-to-dry weight ratio of the tissue before and after perfusion [56]. They also demonstrated that mannitol, a slowly permeating neutral solute, resulted in improved preservation [56]. Furthermore, by adjusting the electrolyte balance (substituting half of the extracellular sodium ions with intracellular ions such as potassium and magnesium), they significantly improved the preservation solution [56]. This alteration boosted the baseline insulin secretion and improved the insulin response to glucose stimulation [56].
Due to the specific challenges linked to ex vivo perfusion of the pancreas, particularly its sensitivity, the duration of ex vivo perfusion for rodent pancreas has been limited to a few hours. One study investigated the effects of leptin on endocrine function by measuring insulin secretion from the isolated pancreas [57]. They demonstrated that leptin does not regulate beta cell function, as the addition of leptin to the system had no effect on carbachol-induced insulin secretion [57]. Other studies measured secretion of pancreatic enzymes (amylase and lipase) in the perfusate, which are indicative of exocrine cell damage [58]. A graphical representation of their EVOP system is shown in Figure 4. In recent years, improvements have been made to pancreas EVOP systems to preserve the tissue for longer periods of time. Since the pancreas is a low vascularized organ and highly susceptible to edema when subjected to high pressures, low-pressure perfusion was introduced [59]. These rodent models contribute uniquely to the study of islet biology but improvements need to be made to extend the organ preservation time, as these studies were limited to short-term maintenance over a few hours.
Figure 4.

Graphical representation of a benchtop rat pancreas EVOP system developed by Mantke et al. [58]. The rat pancreas was isolated and cannulated at the caudal aorta (input) and portal vein (output) using Teflon tubing. The perfusate (modified Krebs solution) was gassed with 95% O2 and 5% CO2 and pumped at a constant flow rate while perfusion pressure was measured by a pressure transducer. A heat exchanger was used to maintain the perfusate at a constant temperature of 37°C. The perfusate was pumped into the cannulated caudal aorta, into the pancreas, and was pumped out of the pancreas through the portal vein, which flowed into a collection bottle, creating an open system.
Recent advances in ex vivo perfusion of pancreas in preclinical models have highlighted the importance of these systems not just for organ preservation and transplantation but also as platforms for disease modeling and studying mechanisms of therapeutic interventions. For instance, numerous studies have used rodent EVOP systems to investigate how the endocrine and exocrine functions of pancreas are impacted by certain conditions (i.e. hypothermia) or therapeutic interventions. These platforms can serve as valuable tools for type 1 diabetes research in assessing how drugs modulate glucose, lactate, and insulin release in physiologically relevant models. Despite progress in ex vivo pancreas perfusion, a major remaining challenge is the risk of edema. Due to its fragile microvasculature and high vascular permeability, the pancreas is highly sensitive to hydrostatic pressure changes [60, 61]. In large animal models, several studies have established low-pressure systems that achieve optimal oxygenation while maintaining tissue integrity. However, further refinement is needed in small animal EVOP models. To combat edema and extend pancreas viability during ex vivo perfusion, addition of a dialysis circuit would be beneficial as it helps remove metabolic waste [60, 62]. Future efforts should focus on developing and optimizing a dialysis circuit to enhance perfusion outcomes.
6. Discussion
Both benchtop and incubator-based EVOP systems have been used widely in small animals for various applications including disease modeling, evaluation of novel therapies, advancing our understanding of organ physiology, as well as regenerative medicine research. Compared to in vivo models, incubator-based and benchtop perfusion systems both allow for more precise control of pressure, oxygenation, and other experimental parameters, and offer real-time visualization of the perfused organ. Benchtop EVOP systems struggle to maintain sterile environments, hence increasing chances of contamination [5]. This significantly limits perfusion of organs for extended periods of time. However, incubator-based EVOP systems minimize the risks of contamination due to the sealed environment of the incubator. Incubator perfusion systems are also more cost-effective, easier to operate, and take up less space. However, both benchtop and incubator EVOP systems face challenges in maintaining physiologic support during prolonged periods of organ perfusion. To remain viable, organs require a continuous supply of nutrients and oxygen, along with constant removal of toxic metabolites. Lack of metabolic regulation in current EVOP systems leads to inflammation, cell death, and organ damage within a short period of time. To address this issue, several technologies have been proposed. For instance, integration of white blood cell filters within the circuit can block leukocyte infiltration, hence mitigating the inflammatory response [63, 64]. Studies show that integration of a cytokine filter during lung [65] and kidney [66] perfusion reduced overall proinflammatory cytokine expression, reduced edema formation, and improved blood flow. This technique has primarily been used in large animal models and humans [67]. While further development is needed for small animal models, it is important to consider the implications of leukocyte depletion on disease modeling and drug development. Another approach to supply the organ with sufficient nutrients and oxygen and remove metabolic waste is through period replacement of the perfusate within the circuit with fresh solution. This method requires large volumes of perfusate, making it expensive. To reduce costs, efforts have been made to recycle perfusate using dialysis-based systems [68]. Finally, xenogeneic cross-circulation is another technique used for enhancing physiologic support during EVOP. First developed by O'Neill et al. in 2017, this technique involves exchanging whole blood between an ex vivo organ and a living organism [69]. Cross-circulation of human donor lungs with swine models has not only extended duration of perfusion [69] but also improved gas exchange and dynamic compliance [70]. Similar to leukocyte filters, this method has only been explored in large animal models and discarded human organs, highlighting the need for further development in small animals.
While these technological advances aim to improve physiologic support during EVOP, system design should also be tailored to the specific functional and anatomical requirements of different organs. For instance, the gut contains both luminal (intestinal lumen) and vascular (blood vessels) perfusion. Luminal perfusion can be used to model intestinal absorption and secretion, as well as studying the epithelium while vascular perfusion is required for achieving sufficient nutrients and oxygen as well as studying the endothelium.
The ex vivo perfusion of lungs also requires additional considerations. To closely mimic physiologic conditions, lungs must be ventilated to simulate lung expansion and contraction [71]. This is especially important for studying normal lung physiology, evaluating therapeutic interventions, as well as preserving lungs for transplantation. To simulate the humid environment of the lungs, researchers have developed methods for wet ventilation which usually involves suspending the lung in media and connecting it to a ventilator [71]. In other cases, dry ventilation can be used to allow for air exchange in the lung, which is closer to physiological and a good environment for disease modeling purposes [46].
Additionally, the kidneys require only vasculature perfusion, but the ureter can be also cannulated to collect urine in order to monitor kidney function. Because of the density of the kidney, optimization of the perfusate flow rate and pressure is important to minimize physical damage but still maintain oxygen and nutrient delivery. The addition of a vasodilator has proven useful in porcine EVOP [72].
The pancreas presents unique considerations for EVOP due to the unique properties of the organ. Being a highly sensitive organ with low flow and pressure, it is difficult to achieve an optimal flow rate. Inadequate flow rates may result in insufficient perfusion, leading to cell death, while too high flow rates can quickly lead to edema and subsequent tissue damage, which has been a challenge for many groups. Furthermore, given its predominantly exocrine composition, the pancreas is particularly prone to the secretion of digestive enzymes, such as proteases and lipases, especially under stress or manipulation. Therefore, the inclusion of enzyme inhibitors, such as trypsin inhibitor, is crucial to prevent autodigestion of the organ.
7. Conclusions
In conclusion, EVOP systems in small animals have enabled researchers to study organ physiology, model complex disease mechanisms, and investigate novel therapeutic agents. EVOP systems in small animals represent a cost-effective yet physiologically relevant model that can significantly progress the field of organ perfusion. Here, we highlighted four organ systems. Although there are common ex vivo parameters that must be met for all organs, such as nutrients, flow rates, and oxygenation, specific organ adaptations such as ventilation for the lung, or electrical nerve stimulation for the heart must be considered. The heart EVOP has a long history that has led to the modern benchtop EVOP systems now used for multiple organs. Bowditch, in 1869, using a frog heart, connected to a cannula and reservoir of rabbit serum, isolated the vagus nerve from the spinal cord to the heart, and was able to stimulate it ex vivo. The Langendorff ex vivo heart system, developed in 1895, revealed the importance of oxygenation to heart function [73]. Modern systems are being used to regenerate acellular organs for organ replacement therapies and evaluate novel pharmaceutical treatments. These systems prove to be valuable models owing to their ability to replicate physiologic conditions in an ethical, cost-effective, and timely manner. Ultimately, small animal EVOP systems offer a powerful and practical platform for advancing medical research and therapeutic discovery.
Funding Statement
This work was supported by the Canadian Institutes of Health Research (CIHR) Foundation Grant 353857 and the New Frontiers Innovations Grant NFRFT-2020-00787.
Contributor Information
Agostino Pierro, Email: agostino.pierro@sickkids.ca.
Ian M. Rogers, Email: rogers@lunenfeld.ca.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Disclosure
All the authors approved the final manuscript.
Conflicts of Interest
The authors declare no conflicts of interest.
Author Contributions
Mina Yeganeh, Andea Zito, Marwa Sadat, and Ian M. Rogers contributed to the research, discussion, writing, and editing of this manuscript. Agostino Pierro and Ian M. Rogers provided advice and supervision. Mina Yeganeh and Andea Zito shared cofirst authorship.
References
- 1.Monbaliu D., Liu Q., Vekemans K., Pirenne J. History of Organ Perfusion in Organ Transplantation. In: Talbot D., D’Alessandro A., editors. Organ Donation and Transplantation after Cardiac Death . Oxford University Press; 2009. pp. 31–50. [DOI] [Google Scholar]
- 2.Cypel M., Yeung J. C., Liu M., et al. Normothermic Ex Vivo Lung Perfusion in Clinical Lung Transplantation. New England Journal of Medicine . 2011;364(15):1431–1440. doi: 10.1056/nejmoa1014597. [DOI] [PubMed] [Google Scholar]
- 3.Mazilescu L. I., Urbanellis P., Kim S. J., et al. Normothermic Ex Vivo Kidney Perfusion for Human Kidney Transplantation: First North American Results. Transplantation . 2022;106(9):1852–1859. doi: 10.1097/tp.0000000000004098. [DOI] [PubMed] [Google Scholar]
- 4.Kumar R., Chung W. Y., Dennison A. R., Garcea G. Ex Vivo Porcine Organ Perfusion Models as a Suitable Platform for Translational Transplant Research: Ex Vivo Porcine Perfusion Models. Artificial Organs . 2017;41(9):E69–E79. doi: 10.1111/aor.12865. [DOI] [PubMed] [Google Scholar]
- 5.Won N., Castillo-Prado J., Tan X., et al. Ex Vivo Perfusion Using a Mathematical Modeled, Controlled Gas Exchange Self-Contained Bioreactor Can Maintain a Mouse Kidney for Seven Days. Cells . 2022;11:p. 1822. doi: 10.3390/cells11111822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Astashkina A., Mann B., Grainger D. W. A Critical Evaluation of In Vitro Cell Culture Models for High-Throughput Drug Screening and Toxicity. Pharmacology & Therapeutics . 2012;134(1):82–106. doi: 10.1016/j.pharmthera.2012.01.001. [DOI] [PubMed] [Google Scholar]
- 7.Lee G., Papapetrou E. P., Kim H., et al. Modelling Pathogenesis and Treatment of Familial Dysautonomia Using Patient-Specific iPSCs. Nature . 2009;461(7262):402–406. doi: 10.1038/nature08320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fine B., Vunjak-Novakovic G. Shortcomings of Animal Models and the Rise of Engineered Human Cardiac Tissue. ACS Biomaterials Science & Engineering . 2017;3(9):1884–1897. doi: 10.1021/acsbiomaterials.6b00662. [DOI] [PubMed] [Google Scholar]
- 9.Stevens L. J., Donkers J. M., Dubbeld J., et al. Towards Human Ex Vivo Organ Perfusion Models to Elucidate Drug Pharmacokinetics in Health and Disease. Drug Metabolism Reviews . 2020;52(3):438–454. doi: 10.1080/03602532.2020.1772280. [DOI] [PubMed] [Google Scholar]
- 10.Xinaris C. Organoids for Replacement Therapy: Expectations, Limitations and Reality. Current Opinion in Organ Transplantation . 2019;24(5):555–561. doi: 10.1097/mot.0000000000000680. [DOI] [PubMed] [Google Scholar]
- 11.Lee S. H., Sung J. H. Organ-on-a-Chip Technology for Reproducing Multiorgan Physiology. Advanced Healthcare Materials . 2018;7(2):p. 1700419. doi: 10.1002/adhm.201700419. [DOI] [PubMed] [Google Scholar]
- 12.Pegg D. E., Fuller B. J., Foreman J., Green C. J. The Choice of Plastic Tubing for Organ Perfusion Experiments. Cryobiology . 1972;9(6):569–571. doi: 10.1016/0011-2240(72)90182-4. [DOI] [PubMed] [Google Scholar]
- 13.Pinezich M., Vunjak-Novakovic G. Bioengineering Approaches to Organ Preservation Ex Vivo. Experimental Biology and Medicine . 2019;244(8):630–645. doi: 10.1177/1535370219834498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Krebs H. A., Henseleit K. Untersuchungen Uber die Harnstoffbildung im Tierkörper. Hoppe-Seyler´s Zeitschrift für Physiologische Chemie . 1932;210(1-2):33–66. doi: 10.1515/bchm2.1932.210.1-2.33. [DOI] [Google Scholar]
- 15.Naddaf Dezfuli S., Huan Z., Mol J., Leeflang M., Chang J., Zhou J. Influence of HEPES Buffer on the Local pH and Formation of Surface Layer During In Vitro Degradation Tests of Magnesium in DMEM. Progress in Natural Science: Materials International . 2014;24(5):531–538. doi: 10.1016/j.pnsc.2014.08.009. [DOI] [Google Scholar]
- 16.Von Horn C., Zlatev H., Kaths M., Paul A., Minor T. Controlled Oxygenated Rewarming Compensates for Cold Storage–Induced Dysfunction in Kidney Grafts. Transplantation . 2022;106(5):973–978. doi: 10.1097/tp.0000000000003854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Termeer C. C., Weiss J. M., Schöpf E., Vanscheidt W., Simon J. C. The Low Molecular Weight Dextran 40 Inhibits the Adhesion of T Lymphocytes to Endothelial Cells. Clinical and Experimental Immunology . 1998;114(3):422–426. doi: 10.1046/j.1365-2249.1998.00729.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. https://www.xvivoperfusion.com/products/steen-solution/STEEN Solution, XVIVO.
- 19.Yang C., Mookerjea S., Nagpurkar A. Clearance of Rat C-Reactive Protein In Vivo and by Perfused Liver. Glycobiology . 1992;2(1):41–48. doi: 10.1093/glycob/2.1.41. [DOI] [PubMed] [Google Scholar]
- 20.Bradford W. L., Nakatsugawa T. Perfusion Analysis of Periportal and Centrilobular Metabolism of Paraoxon in the Rat Liver. Pesticide Biochemistry and Physiology . 1982;18(3):298–303. doi: 10.1016/0048-3575(82)90070-0. [DOI] [Google Scholar]
- 21.Nicholson M. L., Hosgood S. A. Renal Transplantation after Ex Vivo Normothermic Perfusion: The First Clinical Study. American Journal of Transplantation . 2013;13(5):1246–1252. doi: 10.1111/ajt.12179. [DOI] [PubMed] [Google Scholar]
- 22.Fard A., Pearson R., Lathan R., Mark P. B., Clancy M. J. Perfusate Composition and Duration of Ex-Vivo Normothermic Perfusion in Kidney Transplantation: A Systematic Review. Transplant International . 2022;35:p. 10236. doi: 10.3389/ti.2022.10236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lodhi S., Stone J. P., Entwistle T. R., Fildes J. E. The Use of Hemoglobin-Based Oxygen Carriers in Ex Vivo Machine Perfusion of Donor Organs for Transplantation. ASAIO Journal . 2022;68(4):461–470. doi: 10.1097/mat.0000000000001597. [DOI] [PubMed] [Google Scholar]
- 24.Doherty D. H., Doyle M. P., Curry S. R., et al. Rate of Reaction With Nitric Oxide Determines the Hypertensive Effect of Cell-Free Hemoglobin. Nature Biotechnology . 1998;16(7):672–676. doi: 10.1038/nbt0798-672. [DOI] [PubMed] [Google Scholar]
- 25.White C. W., Hasanally D., Mundt P., et al. A Whole Blood–Based Perfusate Provides Superior Preservation of Myocardial Function During Ex Vivo Heart Perfusion. The Journal of Heart and Lung Transplantation . 2015;34(1):113–121. doi: 10.1016/j.healun.2014.09.021. [DOI] [PubMed] [Google Scholar]
- 26.Yang B., Hosgood S. A., Harper S. J. F., Nicholson M. L. Leucocyte Depletion Improves Renal Function in Porcine Kidney Hemoreperfusion Through Reduction of Myeloperoxidase+ Cells, Caspase-3, IL-1β, and Tubular Apoptosis. Journal of Surgical Research . 2010;164(2):e315–e324. doi: 10.1016/j.jss.2010.07.044. [DOI] [PubMed] [Google Scholar]
- 27.Maxey T. S., Enelow R. I., Gaston B., Kron I. L., Laubach V. E., Doctor A. Tumor Necrosis Factor-α From Resident Lung Cells is a Key Initiating Factor in Pulmonary Ischemia-Reperfusion Injury. The Journal of Thoracic and Cardiovascular Surgery . 2004;127(2):541–547. doi: 10.1016/j.jtcvs.2003.09.008. [DOI] [PubMed] [Google Scholar]
- 28.Ahmadipour M., Taniguchi D., Duchesneau P., et al. Use of High-Rate Ventilation Results in Enhanced Recellularization of Bioengineered Lung Scaffolds. Tissue Engineering Part C Methods . 2021;27(12):661–671. doi: 10.1089/ten.tec.2021.0182. [DOI] [PubMed] [Google Scholar]
- 29.Engler A. J., Le A. V., Baevova P., Niklason L. E. Controlled Gas Exchange in Whole Lung Bioreactors: Controlled Gas Exchange in Whole Lung Bioreactors. Journal of Tissue Engineering and Regenerative Medicine . 2018;12(1):e119–e129. doi: 10.1002/term.2408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. PULMODYN Data Acquisition Software, Harvard Apparatus.
- 31. IPL-1 Core Isolated Perfused Lung System, Harvard Apparatus.
- 32.Katsirntaki K., Hagner S., Werlein C., et al. Low-Volume Ex Situ Lung Perfusion System for Single Lung Application in a Small Animal Model Enables Optimal Compliance With Reduction in 3R Principles of Animal Research. Transplant International . 2024;37:p. 13189. doi: 10.3389/ti.2024.13189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nelson K., Bobba C., Ghadiali S., Black S. M., Whitson B. A. Animal Models of Ex Vivo Lung Perfusion as a Platform for Transplantation Research. World Journal of Experimental Medicine . 2014;4(2):7–15. doi: 10.5493/wjem.v4.i2.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bassani G. A., Lonati C., Brambilla D., Rapido F., Valenza F., Gatti S. Ex Vivo Lung Perfusion in the Rat: Detailed Procedure and Videos. PLoS One . 2016;11(12):p. e0167898. doi: 10.1371/journal.pone.0167898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nishiitsutsuji-Uwo J., Ross B., Krebs H. Metabolic Activities of the Isolated Perfused Rat Kidney. Biochemical Journal . 1967;103(3):852–862. doi: 10.1042/bj1030852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hems R., Ross B., Berry M., Krebs H. Gluconeogenesis in the Perfused Rat Liver. Biochemical Journal . 1966;101(2):284–292. doi: 10.1042/bj1010284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ross B., Epstein F., Leaf A. Sodium Reabsorption in the Perfused Rat Kidney. American Journal of Physiology-Legacy Content . 1973;225(5):1165–1171. doi: 10.1152/ajplegacy.1973.225.5.1165. [DOI] [PubMed] [Google Scholar]
- 38.Rosenberger C., Khamaisi M., Goldfarb M., et al. Acute Kidney Injury in the Diabetic Rat: Studies in the Isolated Perfused and Intact Kidney. American Journal of Nephrology . 2008;28(5):831–839. doi: 10.1159/000137683. [DOI] [PubMed] [Google Scholar]
- 39.Hori R., Okamura N., Aiba T., Tanigawara Y. Role of P-Glycoprotein in Renal Tubular Secretion of Digoxin in the Isolated Perfused Rat Kidney. Journal of Pharmacology and Experimental Therapeutics . 1993;266(3):1620–1625. doi: 10.1016/s0022-3565(25)39349-3. [DOI] [PubMed] [Google Scholar]
- 40.Lautenschläger I., Dombrowsky H., Frerichs I., et al. A Model of the Isolated Perfused Rat Small Intestine. American Journal of Physiology-Gastrointestinal and Liver Physiology . 2010;298(2):G304–G313. doi: 10.1152/ajpgi.00313.2009. [DOI] [PubMed] [Google Scholar]
- 41.Lautenschläger I., Wong Y. L., Sarau J., et al. Signalling Mechanisms in PAF-Induced Intestinal Failure. Scientific Reports . 2017;7(1):p. 13382. doi: 10.1038/s41598-017-13850-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yissachar N., Zhou Y., Ung L., et al. An Intestinal Organ Culture System Uncovers a Role for the Nervous System in Microbe-Immune Crosstalk. Cell . 2017;168(6):1135–1148.e12. doi: 10.1016/j.cell.2017.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tan W., Lee G., Chen J.-H., Huizinga J. D. Relationships Between Distention-Butyrate- and Pellet-Induced Stimulation of Peristalsis in the Mouse Colon. Frontiers in Physiology . 2020;11:p. 109. doi: 10.3389/fphys.2020.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Motherwell J. M., Rozenblum M., Katakam P. V. G., Murfee W. L. Bioreactor System to Perfuse Mesentery Microvascular Networks and Study Flow Effects during Angiogenesis. Tissue Engineering Part C Methods . 2019;25(8):447–458. doi: 10.1089/ten.tec.2019.0119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Willi C. E., Abdelazim H., Chappell J. C. Evaluating Cell Viability, Capillary Perfusion, and Collateral Tortuosity in an Ex Vivo Mouse Intestine Fluidics Model. Frontiers in Bioengineering and Biotechnology . 2022;10:p. 1008481. doi: 10.3389/fbioe.2022.1008481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Boyle E. C., Dombrowsky H., Sarau J., et al. Ex Vivo Perfusion of the Isolated Rat Small Intestine as a Novel Model of Salmonella Enteritis. American Journal of Physiology–Gastrointestinal and Liver Physiology . 2016;310(2):G55–G63. doi: 10.1152/ajpgi.00444.2014. [DOI] [PubMed] [Google Scholar]
- 47.Gagliardi M., Monzani R., Clemente N., et al. A Gut-Ex-Vivo System to Study Gut Inflammation Associated to Inflammatory Bowel Disease (IBD) Biology . 2021;10(7):p. 605. doi: 10.3390/biology10070605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gagliardi M., Clemente N., Monzani R., et al. Gut-Ex-Vivo System as a Model to Study Gluten Response in Celiac Disease. Cell Death Discovery . 2021;7(1):p. 45. doi: 10.1038/s41420-021-00430-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Babkin B. P., Starling E. H. A Method for the Study of the Perfused Pancreas. The Journal of Physiology . 1926;61(2):245–247. doi: 10.1113/jphysiol.1926.sp002286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sade R. M. Transplantation at 100 years: Alexis Carrel, Pioneer Surgeon. The Annals of Thoracic Surgery . 2005;80(6):2415–2418. doi: 10.1016/j.athoracsur.2005.08.074. [DOI] [PubMed] [Google Scholar]
- 51.Florack G., Sutherland D. E. R., Heil J., Squifflet J. P., Najarian J. S. Preservation of Canine Segmental Pancreatic Autografts: Cold Storage Versus Pulsatile Machine Perfusion. Journal of Surgical Research . 1983;34(5):493–504. doi: 10.1016/0022-4804(83)90101-4. [DOI] [PubMed] [Google Scholar]
- 52.Ferrer-Fàbrega J., Folch-Puy E., Llaves-López A., García-Pérez R., Fuster J. Breaking the Limits of Experimental Pancreas Transplantation: Working Toward the Clinical Ideal Graft. Frontiers in Transplantation. . 2022;1:p. 1035480. doi: 10.3389/frtra.2022.1035480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Squifflet J.-P., LeDinh H., de Roover A., Meurisse M. Pancreas Preservation for Pancreas and Islet Transplantation: A Minireview. Transplantation Proceedings . 2011;43(9):3398–3401. doi: 10.1016/j.transproceed.2011.09.052. [DOI] [PubMed] [Google Scholar]
- 54.Wahlberg J. A., Southard J. H., Belzer F. O. Development of a Cold Storage Solution for Pancreas Preservation. Cryobiology . 1986;23(6):477–482. doi: 10.1016/0011-2240(86)90056-8. [DOI] [PubMed] [Google Scholar]
- 55.Loubatieres-Mariani M. M., Chapal J., Puech R., Manteghetti M. A Different Action of Hypothermia on Insulin Release From the Isolated, Perfused Rat Pancreas, Depending on the Stimulating Agent. Diabetes . 1980;29(11):895–898. doi: 10.2337/diab.29.11.895. [DOI] [PubMed] [Google Scholar]
- 56.Pegg D. E., Klempnauer J., Diaper M. P., Taylor M. J. Assessment of Hypothermic Preservation of the Pancreas in the Rat by a Normothermic Perfusion Assay. Journal of Surgical Research . 1982;33(3):194–200. doi: 10.1016/0022-4804(82)90029-4. [DOI] [PubMed] [Google Scholar]
- 57.Silvestre R. A., Rodríguez-Gallardo J., Egido E. M., Marco J. Effect of Leptin on Insulin, Glugacon and Somatostatin Secretion in the Perfused Rat Pancreas. Hormone and Metabolic Research . 2001;33:207–212. doi: 10.1055/s-2001-14947. [DOI] [PubMed] [Google Scholar]
- 58.Mantke R., Schubert D., Röcken C., et al. Caerulein or Taurocholate Induced Enzymatic and Histologic Alterations in the Isolated Perfused Rat Pancreas. Langenbeck’s Archives of Surgery . 2009;394(2):363–369. doi: 10.1007/s00423-008-0401-8. [DOI] [PubMed] [Google Scholar]
- 59.Prudhomme T., Renaudin K., Lo Faro M. L., et al. Ex Situ Hypothermic Perfusion of Nonhuman Primate Pancreas: A Feasibility Study. Artificial Organs . 2020;44(7):736–743. doi: 10.1111/aor.13655. [DOI] [PubMed] [Google Scholar]
- 60.Parmentier C., Ray S., Mazilescu L. I., et al. Normothermic Ex Vivo Machine Perfusion of Discarded Human Pancreas Allografts: A Feasibility Study. Transplant International . 2023;36:p. 10936. doi: 10.3389/ti.2023.10936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kervella D., Mesnard B., Prudhomme T., et al. Sterile Pancreas Inflammation During Preservation and After Transplantation. International Journal of Molecular Sciences . 2023;24(5):p. 4636. doi: 10.3390/ijms24054636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Parmentier C., Ray S., Mazilescu L., et al. Normothermic Ex Vivo Pancreas Perfusion for the Preservation of Pancreas Allografts Before Transplantation. Journal of Visualized Experiments . 2022;63905(185) doi: 10.3791/63905. [DOI] [PubMed] [Google Scholar]
- 63.Bruil A., Beugeling T., Feijen J., Van Aken W. G. The Mechanisms of Leukocyte Removal by Filtration. Transfusion Medicine Reviews . 1995;9(2):145–166. doi: 10.1016/s0887-7963(05)80053-7. [DOI] [PubMed] [Google Scholar]
- 64.Iske J., Schroeter A., Knoedler S., et al. Pushing the Boundaries of Innovation: the Potential of Ex Vivo Organ Perfusion From an Interdisciplinary Point of View. Frontiers in Cardiovascular Medicine . 2023;10:p. 1272945. doi: 10.3389/fcvm.2023.1272945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Iskender I., Cosgun T., Arni S., et al. Cytokine Filtration Modulates Pulmonary Metabolism and Edema Formation During Ex Vivo Lung Perfusion. The Journal of Heart and Lung Transplantation . 2018;37(2):283–291. doi: 10.1016/j.healun.2017.05.021. [DOI] [PubMed] [Google Scholar]
- 66.Hosgood S. A., Moore T., Kleverlaan T., Adams T., Nicholson M. L. Haemoadsorption Reduces the Inflammatory Response and Improves Blood Flow During Ex Vivo Renal Perfusion in an Experimental Model. Journal of Translational Medicine . 2017;15(1):p. 216. doi: 10.1186/s12967-017-1314-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Haselwanter P., Scheiner B., Balcar L., et al. Use of the CytoSorb Adsorber in Patients With Acute-On-Chronic Liver Failure. Scientific Reports . 2024;14(1):p. 11309. doi: 10.1038/s41598-024-61658-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wei D., Gao F., Yang Z., et al. Ex Vivo Lung Perfusion With Perfusate Purification for Human Donor Lungs Following Prolonged Cold Storage. Annals of Translational Medicine . 2020;8(3):p. 38. doi: 10.21037/atm.2019.10.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.O’Neill J. D., Guenthart B. A., Kim J., et al. Cross-Circulation for Extracorporeal Support and Recovery of the Lung. Nature Biomedical Engineering . 2017;1(3):p. 0037. doi: 10.1038/s41551-017-0037. [DOI] [Google Scholar]
- 70.Kelly Wu W., Guenthart B. A., O’Neill J. D., et al. Technique for Xenogeneic Cross-Circulation to Support Human Donor Lungs Ex Vivo. The Journal of Heart and Lung Transplantation . 2023;42(3):335–344. doi: 10.1016/j.healun.2022.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bijonowski B. M., Miller W. M., Wertheim J. A. Bioreactor Design for Perfusion-Based, Highly Vascularized Organ Regeneration. Current Opinion in Chemical Engineering . 2013;2(1):32–40. doi: 10.1016/j.coche.2012.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Echeverri J., Goldaracena N., Kaths J. M., et al. Comparison of BQ123, Epoprostenol, and Verapamil as Vasodilators During Normothermic Ex Vivo Liver Machine Perfusion. Transplantation . 2018;102(4):601–608. doi: 10.1097/tp.0000000000002021. [DOI] [PubMed] [Google Scholar]
- 73.Leivaditis V., Mulita F., Dahm M., et al. History of the Development of Isolated Heart Perfusion Experimental Model and its Pioneering Role in Understanding Heart Physiology. Archives of Medical Science . 2024;9(1):109–121. doi: 10.5114/amsad/188270. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
