Skip to main content

Methods of ex vivo analysis of tissue status in vascularized composite allografts

Abstract

Vascularized composite allotransplantation can improve quality of life and restore functionality. However, the complex tissue composition of vascularized composite allografts (VCAs) presents unique clinical challenges that increase the likelihood of transplant rejection. Under prolonged static cold storage, highly damage-susceptible tissues such as muscle and nerve undergo irreversible degradation that may render allografts non-functional. Skin-containing VCA elicits an immunogenic response that increases the risk of recipient allograft rejection. The development of quantitative metrics to evaluate VCAs prior to and following transplantation are key to mitigating allograft rejection. Correspondingly, a broad range of bioanalytical methods have emerged to assess the progression of VCA rejection and characterize transplantation outcomes. To consolidate the current range of relevant technologies and expand on potential for development, methods to evaluate ex vivo VCA status are herein reviewed and comparatively assessed. The use of implantable physiological status monitoring biochips, non-invasive bioimpedance monitoring to assess edema, and deep learning algorithms to fuse disparate inputs to stratify VCAs are identified.

Introduction

Vascularized composite allotransplantation involves the transfer of anatomical structures containing multiple tissue types including skin, bone, fat, muscle, and connective tissue from one individual to another. Vascularized composite allografts (VCAs) range from face and hand to less commonly transplanted grafts such as the abdominal wall, uterus, and penis [1]. To date, more than 120 upper extremity and 46 face transplants have been performed worldwide along with other types of VCAs [2, 3]. Successful short- and long-term transplantation outcomes demonstrate VCA as viable treatment option for patients suffering large tissue defects or loss of limbs and for whom there are no conventional reconstructive options [4,5,6,7]. Currently, around 185,000 amputations take place in the US every year due to trauma, oncological resection, or severe burn, with the total number of amputees expected to reach 3.5 million by 2050 [8]. However, high immunogenicity and antigenicity are significant challenges in VCA, particularly with recipient response to skin. Acute rejection rates are approximately six-fold greater in VCA than in solid organ transplantation, necessitating treatment with immunosuppression and, in some instances, result in graft loss [1]. For amputees, limb prosthetics serve as an alternative to VCA transplantation. While prosthetics avoid the immunological challenges of graft rejection, functional and sensory recovery are unparalleled in VCA transplantation. Furthermore, prosthetics contribute to a significant rate of rejection as they are largely limited in practical applications, difficult to maintain, and awkward or uncomfortable to use [9]. While generally non-lifesaving, vascularized composite allotransplantation significantly improves an amputee’s quality of life, enables full functional recovery, and substantially recovers potential economic productivity loss [3, 10, 11].

Orchestrating the logistics of preserving and transporting the tissue remains a significant barrier to VCA accessibility. Once the graft is removed from circulation, a pathophysiological signaling cascade initiates due to ischemia and storage on ice. Under cold ischemia, grafts develop inflammatory signaling, muscle necrosis, mitochondrial dysfunction, and degraded vascular integrity [12,13,14]. Herzberg et al. and Piza-Katzer demonstrated that the extent of ischemic injury is inversely correlated with graft function in a bilateral hand transplantation study [15, 16]. Using the current gold standard method of static cold storage, the window of time for VCA viability is approximately 4–6 h [17]. This restricted time frame limits the geographic proximity of VCAs available for transplantation [18]. Compounding this constraint, strict aesthetic and anatomical features are necessary. While human leukocyte antigen (HLA) status is a metric for both organ and VCA suitability, anatomical criteria such as bone size, skin color, and soft tissue features are considered in VCA [19]. Prolonging the viability of VCAs beyond current constraints would therefore expand the availability of compatible allografts.

Advancing machine-perfusion-based preservation

The gold standard in organ preservation, static cold storage, involves inducing hypothermia to reduce the metabolic demand of an organ or graft. Upon procurement, the VCA is flushed with a cold preservation solution and maintained on ice during transport. During this time, oxygen deprivation and the switch to anaerobic metabolism contribute to ischemic tissue injury [20]. In lowering the preservation temperature, static cold storage preserves the graft by slowing cellular metabolic activity [21, 22]. Despite its simplicity and widespread adoption in the clinic, static cold storage beyond a few hours is associated with early graft dysfunction in VCA due to tissue injury [20,21,22]. Muscle and nerve are particularly relevant to VCA preservation as these tissues are highly susceptible to ischemic damage [22,23,24,25]. In addition, once circulation is re-established and oxygen re-introduced to the now ischemic tissue, damage is exacerbated by reperfusion injury, a complex, injurious, pathophysiological cascade [13, 23, 26]. A number of detailed reviews on the molecular and cellular underpinnings of reperfusion injury have been published [12, 25, 27, 28]. Briefly, upon re-establishing blood flow, oxidative stress forms reactive oxygen species (ROS). Once the graft is transplanted, ROS and inflammatory signaling aggravate the innate immune system of the VCA recipient, leading to a cascade of tissue damaging events that encompass acute rejection. Furthermore, a study by Friedman and colleagues emphasized the contribution of injury-induced inflammation in acute allograft rejection [29]. Recently, machine perfusion has emerged as an alternative to ischemia that mitigates the consequences of metabolite accumulation and anoxia.

Ex vivo perfusion

Over the last few decades, ex vivo machine perfusion has presented promising options for prolonging organ preservation time. Figure 1 illustrates the design and implementation of a generalized machine perfusion bioreactor used in VCA. The intent of perfusion is to reduce the extent of ischemic damage by removing harmful metabolites while delivering nutrients to sustain cellular metabolism [30]. As a well-established focus in transplant medicine, detailed reviews of organ preservation solutions and their compositions have been published [31, 32]. Generally, perfusates include the following components: colloids to minimize edema by increasing oncotic pressure, ions to preserve membrane function or serve as buffering agents, saccharides to sustain glycolysis and/or increase osmotic pressure to counteract edema, and dilute red blood cells or artificial hemoglobin to facilitate oxygen transport [30, 31, 33]. Furthermore, sodium bicarbonate is included to balance metabolite accumulation and insulin is included to increase glucose uptake [17, 20, 34]. Perfusion typically occurs through attachment to intact arteries and vessels to pulsatile perfusion systems following [18]. In early studies, pumping perfusate through grafts led to significant pressure-induced tissue injury, or barotrauma [35]. Nonetheless, there has been noteworthy progress and even commercialization of ex vivo perfusion systems designed for solid organs including the kidney, liver, lung, and heart [36]. Much research over the last 30 + years has focused on adapting solid organ perfusion systems for VCA preservation [37,38,39,40,41]. In experimental VCA, perfusates adapted from solid organ transplantation such as STEEN Solution™, Perfadex®, Custodiol® HTK, Celsior®, and University of Wisconsin (UW) Solution have been used to perfuse tissue with comparable efficacies [42,43,44,45]. Blood-based perfusates have also been used to facilitate oxygen delivery [41, 46]. Promising outcomes from large animal models encouraged Werner and colleagues to study ex vivo perfusion in human VCA, subsequently demonstrating the feasibility of human limb machine perfusion for 24 h with plasma-based hemoglobin [47]. In a surgical setting, VCA is typically perfused prior to static cold storage with UW solution established as the preferred perfusate. However, the empirical rationale for its selection has not been thoroughly characterized [30, 48]. While differences in transplant outcomes have been compared in solid organ preservation, comparative data in VCA is limited and there currently is no consensus on optimal perfusate composition [49,50,51,52,53]. Despite advances in VCA perfusion methodology, corresponding metrics to evaluate their efficacy need to be identified in order to catalyze advancement of the field.

Fig. 1
figure 1

© 2017 Wolters Kluwer Health)

Perfusion bioreactor design and implementation. The general layout of the machine extracorporeal perfusion system commonly used in VCA ex-vivo perfusion. A A schematic illustration of the perfusate circulation as a system comprising a the porcine forelimb in the perfusion box on top of a b metal grid that allows c passive venous drainage of the preservation fluid, d a 15 mm needle probe that measures muscle temperature, e collection reservoir, f a centrifugal pump, regulating in-line pressure at ≤ 30 mmHg, g a membrane oxygenator, infusing the fluid with a mix of 95% O2 and 5% CO2, h heater-cooler machine, cooling the fluid to 8–10 °C, i drug administration point/fluid sampling port, and j flowmeter. (Image reproduced with permission from [54] ©2020 Anne Sophie Kruit et al. Transplant International published by John Wiley & Sons Ltd on behalf of Steunstichting ESOT.) B An actual perfusion system showing a human arm in the bioreactor chamber. C and D Photos of actual perfusion bioreactor systems consisting of an allograft housing, perfusate pump, perfusate oxygenator, heater, sampling port, and flow meter. (Images reproduced with permission from [47]

Surrogate biomarkers of VCA injury during preservation

Within the complex tissue architecture of VCA, nearly imperceptible damage occurs during preservation. Vascular degeneration, necrosis, and metabolite accumulation include those changes which are not visually detectable yet lower the likelihood of successful transplant outcomes. Evaluation of tissue integrity and pathology is typically achieved with tissue biopsy and subsequent histology. Using this method, features such as cellular muscle structure and edema can be visualized with high resolution. The Banff Criteria ranks tissue pathology in VCA and encompasses a set of histopathological features that distinguish severity by the extent and localization of immune infiltrate (Table 1) [55, 56]. Enhancing its resolution, Rosales and colleagues have expanded on the Banff Criteria to stratify epidermal and vascular tissue features [57]. Immunostimulatory molecules evoked by surgical tissue damage and altered metabolic activity under ischemia trigger the infiltration of macrophages into muscle and skin [56, 58]. While the resolution of tissue histology may yield invaluable insight for VCA status, applications to ex vivo graft preservation are limited due to its invasiveness, time-intensiveness, intrinsic inter- and intra-observer variability, and lack of quantification. In one center, it was found that approximately 80% of in vivo skin containing VCA rejection episodes that were resolved with topical therapy or oral immunosuppression were scored as Grade III using the Banff Criteria [59]. Interestingly, VCA with rejection episodes necessitating systemic administration of immunosuppression were also scored as Grade III. To overcome the uncertainty associated with histopathological grading, quantifiable VCA features and alternative biomarkers of viability have emerged in recent time. A review of the literature that employs a range of tissue monitoring methods was conducted using Scopus and PubMed. Given the relatively recent development of VCA surgery, all original research articles up to the present were included. Search terms included ‘vascularized composite tissue allograft/allotransplantation’ in combination with the following terms: biomarker, monitoring, diagnostics, perfusion, viability, rejection, ex vivo preservation.

Table 1 Banff classification of acute rejection in skin-containing allografts

Figure 2 provides an illustration that summarizes the various methods of analysis of tissue status during preservation of allografts in VCA. The illustration follows the methods described in the following sections.

Fig. 2
figure 2

© 2015 Royal College of Ophthalmologists, 2019 American Chemical Society, respectively.)

An illustrative summary of methods of analysis of tissue status during preservation of allografts in VCA. Clockwise: Metabolic, Biochemical, Histopathological, and Biophysical. (Images reproduced with permission from [60, 61]

Metabolic and biochemical biomarkers

During solid organ perfusion, cellular metabolites are often used as surrogates of transplant viability. With effective machine perfusion, perfusate carries biomolecules from the interstitial space for sample collection and analysis. These surrogate viability markers are derived from normal metabolic processes, whose deviation from established physiological values are thought to indicate graft deterioration. Once the VCA is removed from the donor, the surgical tissue damage, the ischemic period and lack of circulation alters the graft on a molecular and cellular level. Oxygen and nutrient deprivation lead to series of ionic and metabolic changes in cells over time. Membrane potential and ion compartmentalization can be maintained with low ATP reserves early in ischemia, however, after a few hours, mitochondrial activity is altered and membrane potential decreases [14]. Without blood flow, cells are still able to synthesize ATP through anerobic metabolism and phosphocreatine (PCr) pathways, such that ATP drops slowly as PCr and glycogen remain abundant. However, three hours into ischemia, ATP declines quickly, followed by complete consumption of PCr and glycogen 6 h post ischemia [62]. The switch from aerobic to anaerobic metabolism has implications in metabolite accumulation that may give insight to graft viability and functionality. Upon ischemia, the change in cellular metabolism result in the accumulation of NAD, lactate, and H+, thus lowering intra- and extracellular pH [23]. Furthermore, lactate, creatine kinase, potassium, lactate dehydrogenase have been identified as indicators of ischemia [63]. Lactate concentration is commonly used as a surrogate marker of cellular stress and hypoxia in solid organ transplantation and is often used as a marker for VCA viability [20, 43, 64, 65]. Lactate above a concentration of 5 mmol/L at the end of the heart perfusion has been suggested as an indicator of poor post-transplant outcomes [66]. In a human limb tourniquet study, lactate reportedly increased to 28 mmol/L after 36 min of ischemia, subsequently 15 mmol/L has been suggested as initial estimate for VCA lactate threshold [67]. Acidosis is associated with reduced VCA functionality for ex vivo timepoints, specifically in studies which use muscle contractility as another metric of viability [68]. Additional study of the correlation between pH and VCA outcome is needed to bolster biomarker efficacy.

Concurrently, glucose, phosphocreatine, and ATP content of tissue inform its rate of energy consumption. As graft viability deteriorates, metabolism is expected to slow to an eventual stop. Liquid chromatography-mass spectrometry (LCMS) as well as blood-gas analysis have been used to quantify metabolites contained in tissues or perfusate following static cold storage or perfusion. Compared to a warm treatment group in a rat hindlimb model, Gok and colleagues demonstrated that static cold storage incurred the greatest decrease in phosphocreatine and creatine using LCMS [22, 69]. However, a significant difference in ATP content or the ratio of ATP/ADP was not evidenced in either study conducted. In another study, similar findings indicated a non-statistically significant increase in glucose content of cold stored VCA after 6 h of perfusion [20]. More recently, in-line glucose sensors have been applied to organ perfusion to continuously monitor perfusate [70]. Despite some encouraging evidence of glucose as a metric of cellular metabolism, it may be challenging to use as a biomarker when added to perfusate. This issue necessitates the use of more sensitive glucose sensors as well as precise control of perfusate composition.

Calcium is a key mediator of muscle contraction. During tissue and organ preservation, ischemia induces mitochondrial dysfunction and alters cellular calcium retention [71,72,73,74,75]. This process has been characterized in cardiac transplantation and is associated with impaired organ function [71, 76]. In VCA, metabolically-active skeletal muscle is highly susceptible to ischemic injury. Depletion of ATP inhibits ion exchange by sodium–potassium ATPases as well as calcium ATPases [72, 75]. Consequently, sodium-calcium antiporters reverse in mechanism to restore cytosolic sodium concentration, leading to an accumulation of cytosolic calcium [72]. Sodium, calcium, and potassium concentration are often used as a measure of ischemia in VCA. Specifically, increased intracellular ion retention has been used to identify the onset of tissue ischemia during preservation. In a study by Amin and colleagues, porcine forelimbs were preserved under an extended period of static cold storage or perfusion with varying temperatures and mean arterial pressures for a total of 8 h prior to transplantation and reperfusion [20]. During reperfusion, blood-gas analysis revealed a destabilized electrolyte profile and acidosis in the static cold storage group, this was treated with repeat infusions of bicarbonate to prevent graft loss. These limbs demonstrated depleted sodium and calcium levels and increased potassium relative to the machine perfusion groups.

The consequences of tissue damage due to ionic metabolite imbalance are not well-understood. Despite an apparent connection between VCA viability and metabolic activity, measurement of biochemical parameters is confounded by the lack of standardized methods in preservation. Differences in graft model, ischemic duration, perfusate composition, and perfusion parameters such as flow rate and pressure may influence the accumulation of metabolites. In machine perfusion systems, metabolite accumulation in the perfusate may indicate the status of cellular metabolism and severity of ischemic injury. Perfusate choice may also affect metabolite measurement yet has not been comparatively assessed in VCA. In one study, the impact of perfusate composition was shown in relation to lactate abundance. Kruit and colleagues demonstrated lactate level in abdominal wall machine perfusion perfused by UW was higher than tissue perfused with Custodiol®HTK, which may empirically reflect the intended formulation of UW to support cellular metabolism, whereas HTK is designed to stabilize membrane potential and minimize cell activity [77]. In order to use cellular metabolites as surrogates of tissue injury, further research should consider contributions of perfusate composition and biophysical parameters to quantified tissue status. The forgoing suggests that continued development of fully intra-muscularly indwelling physiological status monitoring biochips that measure metabolites such as glucose, lactate, pH (acidosis) and potassium promises to be welcomed additions to the VCA metabolite monitoring arsenal [78,79,80,81,82,83,84].

Biophysical biomarkers

Once grafts are removed from the donor and subject to static cold storage or perfusion, the biophysical properties of composite tissues become substantially altered. In organ transplantation, donor history including age, illness, medication regimen, and cause of death have been used to confirm acceptability. Studies applying machine perfusion to organs from non-heart-beating donors (NHBD) indicate perfusion pressure and vascular resistance as markers of graft status and perfusion efficiency. In a study of NHBD kidneys, pulsatile hypothermic perfusion precedes a reduction in renal vascular resistance, improved cortical flow, and clot ejection from microcirculation [20, 22, 68, 85, 86]. These observations support that machine perfusion enabled effective perfusate delivery. In a similar study, ischemic kidneys had an elevated vascular resistance (1.25 mmHg/mL/min) compared to a perfused group (0.75 mmHg/mL/min) [87]. Furthermore, warm ischemia time has been shown to increase the risk of delayed graft function in NHBD organs, which may be due to acute tubular necrosis [85]. Vascular resistance in NHBD kidneys has also been investigated as prognostic factor of transplantation outcome in combination with donor history parameters. Kidneys transplanted with low vascular resistance after 20–30 h of hypothermic perfusion with UW have shown improvement in graft survival comparing to average NHBD kidneys survival [88, 89]. Intra-organ vascular resistance at the beginning of perfusion has been demonstrated to directly correlate with warm ischemia time, which may be mitigated by prolonging machine perfusion prior to transplantation [90].

Under native physiological conditions, the endothelium maintains vascular homeostasis. The elasticity of intact blood vessels and capillaries facilitates an adaptive response to varying hemodynamic pressures [91]. As a physiological analog to electrical circuits, vascular resistance is the opposition to hemodynamic flow [92]. Generally, organ machine perfusion systems follow pressure-controlled protocols. The lower end of physiological blood pressure has been shown as optimal to prevent tissue edema [17, 93]. This parameter is influenced by vessel dimensions, perfusate viscosity, flow rate, and temperature [20, 41, 50, 91]. Additionally, perfusate colloid concentration has been shown to affect vascular resistance. In kidneys perfused with a varying colloid concentrations, vascular resistance was observed to be higher in kidney perfused with lower colloid perfusate in comparison with perfusate containing higher colloid [94]. Change in vascular resistance is often used as a surrogate for perfusion efficacy and vascular integrity. Whereas decrease in resistance may occur due to barotrauma and vascular collapse, stable resistance may indicate intact vasculature. During extended limb perfusion with a pressure-controlled circuit, vascular resistance tends to decrease within an hour of perfusion and stabilize thereafter [34, 43, 47]. Gok and colleagues demonstrated that a perfusion flow rate of approximately 1 mL/min lead to a gradual increase in perfusion pressure, barotrauma, lactate, and limb edema [43]. In another instance, a perfusion circuit by Fahradyan and colleagues increased flow rate until a physiological arterial pressure of 102.9 ± 1.76 mmHg was reached [68]. At the end of perfusion, vascular resistance was found to be increased by 6.42% ± 18.41%. The authors posit that the observed increase in vascular resistance was due to vascular spasm, endothelial edema, and microvascular collapse leading to perfusion failure [68]. Generally, regardless of temperature, perfusate and organ intra-organ vascular resistance during long-term perfusion has been recorded as high at the beginning of machine perfusion but declines and stabilizes as perfusion continues beyond 1 h. Due to differences in experimental conditions and perfusate composition, a significant change in any of these parameters during perfusion may impact pressure reading [95]. Global tissue physiology and comparative analysis of perfusate parameters must be considered in the evaluation of vascular resistance as a surrogate for tissue status.

Aside from ensuring tissues are viable, they must also be functional for successful VCA transplantation outcomes. Due to the accumulation or imbalance of key ions and metabolites, muscle tissue contractility and graft function deteriorate [47, 69, 86]. Namely, allografts such as the face and limbs must enable muscular capabilities in the recipient. Ex vivo VCA tissue assessments attempt to validate functionality by testing single-muscle contractility, where a decay in force generated by the muscle fiber is a sign of reduced graft function. This measurement is often taken by excising and permeabilizing single muscle fibers, then placing them in a calcium-containing solution. Single fibers are dissected then secured to a force transducer. Some researchers have assessed contractional force by immersing tissue in solutions containing high concentrations of calcium and ATP [47, 86]. Alternatively, muscle contraction has also been induced by using point electrodes to deliver a supramaximal electrical stimulus to whole ex vivo limbs [22, 69]. The force generated is normalized to the cross-sectional area of the muscle and used to estimate fiber contractility and functionality. Constantinescu and colleagues found that perfused porcine forelimbs maintained a motor response to electrical stimulation for the duration of 12-h perfusion, whereas non-perfused tissue was unresponsive after 30 min [41]. Across experiments comparing muscle contractility in static cold stored and perfused grafts, the latter has been shown to sustain muscle contraction for up to a maximum of 12 h [69, 86]. Furthermore, it was found that reattachment of static cold stored limbs did not recover force generation, indicating that perfusion may be better suited for long-term ex vivo VCA preservation [86]. A correlation between the duration of cold ischemia on limb function has been investigated by Tsuji and colleagues, who histologically identified muscle degeneration and necrosis in rat hind limbs maintained at 4 °C for up to 72 h [96]. The findings were validated using electromyography 3 weeks following transplantation, which indicated a longer delay in motor response for all ischemic groups compared to limbs transplanted immediately. In a separate functional validation, one group used porcine abdominal muscle flaps with electrical field stimulation used for recording muscle contraction [97]. This inherently non-contact electrical field stimulation approach may be better suited for graft transport during ex vivo preservation [41, 97]. As a whole, muscle contractility is surrogate for graft function that may inform long-term transplant success, although factors such as preservation condition, ischemic and/or reperfusion damage, and the integrity of nerves and muscles must be considered with tissue force generation to identify causes of reduced tissue functionality. The continued development of multiplexed multielectrode bioimpedance spectroscopy used in the measurement of edema, with and without electric field stimulation, may be a welcomed addition for the continuous monitoring and stratification of VCA status during preservation [98,99,100,101].

Histopathology and tissue composition biomarkers

Aside from biochemical and biophysical VCA components, additional factors within the tissue may inform graft viability. Markers of inflammation such as edema, cytokine profiles, and immune cell subsets have been explored as potential biomarkers in VCA. In skin-containing allograft preservation and transplantation, edema is commonly used as an indication of graft injury or rejection [55, 57, 102,103,104]. Typically, during experimental VCA, samples of tissue are isolated then desiccated, the initial “wet” weight and subsequent “dry” weight yield a ratio used as a surrogate metric of edema [105]. Alternatively, histology is commonly used to increase the resolution of tissue assessment and identify edema on a cellular level. Figure 3 is a collage illustrating the significance of edema as a clinical indicator of acute rejection in VCA. In a porcine skin-containing VCA flap model, Rosales and colleagues used histology to determine extent of inflammation, as evidenced by immune infiltrate and edema [57]. From transplant recipients of either haploidentical or class I MHC mismatched groups, 28 serial biopsies were taken from 8 animals and ranked by the relative amount of inflammatory infiltrate for correlation to graft survival time. Biopsies from the mismatched transplant indicated a greater presence of immune cells as well as marked edema compared to the matched group where mild edema was noted. The MHC mismatched transplants were ultimately rejected. While the scoring methodology delineated relative degrees of inflammatory infiltrate, the correlation of edema with graft survival lacks quantitative rigor. Therefore, the extent of edema across the two groups was difficult to associate with graft survival. In a swine hindlimb transplantation model, Etra and colleagues applied the Banff grading system to characterize skin rejection [55]. Grafts that exhibited a pattern of severe edema, erythema, and local inflammation were stratified for abandonment rather than treated with immunosuppressive therapy. This was due to the purported low feasibility of graft rescue. Furthermore, the authors noted that the Banff grading criteria was not sufficient for categorizing the range of graft conditions observed. Additionally, some edematous transplants recovered from mild tissue swelling, thus indicating that more quantitatively rigorous VCA stratification may be necessary [106]. Quantifying the extent of edema may identify a definitive correlation between tissue swelling and graft outcomes, thereby expanding the toolkit of biomarkers applicable to ex vivo VCA [99, 100]

Fig. 3
figure 3

Edema is the main Indicator of acute rejection in VCA. A Representative images of edema manifestation in face allograft acute rejection i. No rejection (POM21), ii. early rejection (POM 8) and iii. late rejection (POM26). (Images reproduced with permission from [2] © 2019 Mary Ann Liebert, Inc.) B Edema manifestation in hand allograft acute rejection (Images reproduced with permission from [107] © John Wiley & Sons, Inc.) C Skin allograft acute rejection graded based on Banff Scoring System. Normal skin: unaffected skin, GradeI: mild perivascular infiltration, GradeII: mild perivascular infiltration with/without mild epidermal or adnexal involvement. No epidermal dyskeratosis or apoptosis, Grade III: dense inflammation and epidermal involvement with apoptosis, dyskeratosis, and/or keratinolysis, Grade IV: necrotizing acute rejection. necrosis of skin structures. (Images reproduced with permission from [108] © 2013 Ravi Starzl et al.) D Association of clinical signs or subtherapeutic tacrolimus levels with acute rejection episodes. (Image reproduced with permission from [2] © 2019 Mary Ann Liebert)

In solid organ transplantation, in vivo biomarkers have been explored as surrogates to predict long-term survival [109,110,111,112]. Components present in blood or serum are often targets for indirect biomarker discovery due to the inherent challenge of sampling from internal organs. For example, blood accumulation of myoglobin, a protein that binds oxygen to facilitate its diffusion in muscle, has been used as a surrogate for cardiac and skeletal muscle injury [113, 114]. In VCA, limited studies have explored non-invasive biomarkers for tissue viability [115]. Honeyman and colleagues have described the following biomarkers identified in VCA: T cell infiltration, T cell effector molecules, and mRNA transcripts of genes associated with rejection [115]. Similarly, Puscz and colleagues have assessed histological and immunological indicators in their study of rat hindlimb transplantation [116]. Chronic rejection (CR) in grafts was identified as the appearance of erythema or edema and subsequently rescued with cyclosporine A and dexamethasone. Compared to non-immunosuppressed rats and those receiving constant cyclosporine A, the CR group exhibited intimal hyperplasia with an approximate three-fold increase in proliferation of intimal cells within the femoral artery. Additionally, immunohistochemistry (IHC) revealed an increased accumulation of CD4+ and CD68+ cells in the CR group. Gene expression analysis of interleukin 6 (IL6), tumor necrosis factor (TNF), and interferon gamma (IFNG) indicated a statistically significant increase in IFNG. In cytokine microarrays, upregulation of various interferon genes was observed in the CR group. In a similar manner, proteomics in chronic rejection has been explored in vivo. Kollar and colleagues evaluated longitudinal serum samples from face transplant recipients to identify potential proteins of interest [117]. The following were found to be highly abundant in instances of severe rejection: MMP3 (Matrix Metalloproteinase 3), ACY1 (Aminoacylase-1), IL1R2 (Interleukin-1 receptor type 2), SERPINA4 (Kallistatin) and CPB2 (Carboxypeptidase B2). As an advantage to IHC or biopsy, proteomic analysis enables non-invasive detection of potential markers of tissue status. The novelty of using proteomic discovery tools in VCA biomarker research presents a promising non-invasive method to evaluate tissue status in vivo, and potentially during ex vivo preservation.

Circulating donor-derived DNA and microRNA is another direction for biomarker development that has been explored in solid organ transplantation and in vivo VCA [118,119,120,121]. When cells and tissues of the donor allograft degrade in vivo, fragmented nucleic acids release into the bloodstream and are cleared within 30 min to two hours. The resulting cell-free donor DNA can be detected in blood or urine [121,122,123]. In a preliminary study, Haug and colleagues quantified cell-free donor-derived DNA from plasma samples from a face transplant recipient as well as a bilateral arm transplant recipient [124]. The accumulation of cell-free donor-derived DNA was correlated with rejection, although its increased concentration was not solely dependent on rejection onset. While measuring cell-free DNA is non-invasive and quantitative, detection sensitivity and enhanced understanding of cell degradation in transplanted VCA is still under refinement. In infectious disease and cancer, microRNAs (miRNAs) have long been a source of interest as potential diagnostics [125,126,127,128]. In particular, circulating nucleic acids such as miRNAs can serve as evidence for pathology due to its highly specific and unique signature [129]. Recently, Di Stefano and colleagues have reviewed miRNAs identified in VCA for the detection of allograft rejection [130]. Various miRNAs with immunomodulatory or effector roles were described with their relation to incidence of rejection. In one study, graft deterioration due to ischemia was discussed in the context of myocardial injury, however a correlation was not observed for the prognostic miRNAs identified. Despite significant developments in miRNA applications to solid organ transplantation, comparable progress in VCA is lacking [131,132,133]. Nonetheless, the prognostic modality has potential for limited applications in ex vivo tissue assessment such as screening for immunogenicity and ischemic damage.

Non-invasive VCA evaluation methods

Clinically relevant avenues to expand upon the applicability of VCA biomarkers include spatial capability, continuous feedback, and real-time quantitation. Medical imaging technology confers these properties and has been applied to ex vivo and in vivo VCA analysis. Conventional methods including x-ray, computed tomography (CT), magnetic resonance imaging (MRI), and high-resolution ultrasound have been leveraged to visualize tissues on a cellular level. An increased resolution in VCA monitoring has been used to reveal dynamic processes such as microcirculation and perfusion, relative change in cellular metabolism, and the development of pathophysiology. These methods have been used primarily for peri-operative monitoring of in vivo VCA [134]. Nonetheless, medical imaging presents advantages to existing VCA biomarkers and has the potential for widespread clinical utility, particularly for monitoring tissues during preservation (Table 2).

Table 2 A comparison of clinically relevant features of VCA assessment modalities

Imaging in the infrared and visible spectrum

In VCA transplantation, vascular patency and occlusion are parameters relevant to graft failure as perfusion is associated with microcirculatory damage [135]. For this reason, methods to assess VCA tissue leverage flow dynamics to quantify microcirculation [136, 137]. Laser Doppler imaging (LDI) has been used to dynamically visualize vasculature, blood flow, and perfusion in superficial tissues [138, 139]. In a rat hind limb free flap transplant model, the method has been used to visualize tissue perfusion to diagnose vascular stenosis in VCA in vivo. [140]. Similarly, thermographic imaging has also been used for the noninvasive, non-contact detection of vascular disorder and inflammation in vivo. [68, 141, 142]. It has also been used to monitor limb temperature during perfusion to verify normothermia [68]. Recent advancements in hardware have simplified thermographic measurement to enable rapid data acquisition using smartphone-compatible technology [143]. While vascular integrity and neo-angiogenesis are relevant markers in vivo, pre-transplantation biomarkers of VCA remain unestablished. In order to elucidate fluid dynamics of VCA during preservation, near-infrared detection methods have been used to develop models of pathology [139]. Building upon optical technology, near-infrared (NIR) methods including ICG lymphoscintigraphy and lymphangiography enable deeper light penetrance into tissue [139, 144]. For example, ICG lymphangiography can be used to track real-time fluid movement in VCA for the quantification of lymphatic drainage [145, 146]. Lymphangiography involves administration of contrast agents and requires high technical skill to accomplish delivery via cannulation. Similarly, fluorescence angiography with indocyanine green (ICG) has been used to visualize microcirculatory and peripheral perfusion in VCA [68, 147]. Although fluorescence imaging has high diagnostic sensitivity, signal attenuation in deeper tissues may occur due to limited light penetrance and scattering [139, 148, 149]. Furthermore, ICG can bind proteins present in the fluid, leading to reduced image resolution [146]. Nonetheless, this method non-destructively enables real-time visualization of fluid circulation in ex vivo models of VCA, which may be used to indicate perfusion efficacy.

Imaging methods that use visible light are semi-quantitative and have relatively minimal hardware requirements. Optical 3D scanning using camera-based measurement has been applied experimentally for the quantification of leg edema in vivo [150]. Parameters including leg curvature and instep height can be derived from 3D coordinates in order to correlate measurements to fluid retention. However, confounding factors such as patient activity and measurement error based on selected reference points reduce reliability (r = 0.64). Optical coherence tomography (OCT) increases the imaging resolution to a micrometer scale and is commonly used to detect macular edema [151,152,153]. In soft tissue, OCT has been applied to temporally quantify edema in a mouse ear burn model [145]. However, due to the use of visible light, this method has low penetrance of non-opaque tissues [145]. While OCT has been demonstrated in macular and auricular tissues to detect interstitial fluid accumulation, this method currently has not been demonstrated for use in VCA.

X-ray and computed tomography

Under appropriate contrast enhancement, X-ray and computed tomography (CT) enable visualization of soft tissue features such as blood vessels, blood clots, and tumors [154]. MicroCT (µCT) is an application of X-ray imaging with miniaturized hardware and enhanced resolution. The technology is commonly used to visualize vasculature in high resolution and has been used in ex vivo VCA, namely to visualize tissue perfusion and pressure-induced damage and to inform perfusion regimens during preservation [155]. While µCT imaging can measure surrogates of transplantation success, such as neo-vascularization, its utility in ex vivo VCA has not been thoroughly established. Furthermore, long data acquisition times of multiple hours for dense tissue, limits its applicability in VCA preservation. Additionally, evaluating cellular and molecular tissue details requires the injection of molecular agents to increase soft tissue contrast [139, 156]. Despite its current limitations, the high resolution spatial information provided by µCT yields prognostic value in a preservation setting and the miniaturized technology is deployable for ex vivo monitoring.

Magnetic resonance imaging

Magnetic resonance imaging (MRI) provides high spatial resolution and extensive visualization of soft tissue [157]. In T2-weighted images, edema can be detected in soft tissues using standardized procedures [158, 159]. Subsequently, T2-weighted methods can assess edema for VCA in vivo. [160, 161]. Beyond traditional MRI, functional MRI (fMRI) techniques reveal dynamic processes. This method has been used to obtain spatiotemporal information in kidney transplantation, myocardial infarction, and cerebral tumors for edema detection [160, 162, 163]. In VCA, the functional MRI method of flow MRI has been used for longitudinal follow-up of transplant vascularization by measuring intravascular flow [164]. In contrast to X-ray or CT methods, flow MRI can be used to evaluate hemodynamics in the absence of an injected contrast agent, thereby minimizing shortcomings associated with uptake of contrast agents by tissue. Alternatively, blood-oxygen-level-dependent (BOLD) fMRI has been presented as a novel, noninvasive alternative to oxygen-sensing electrodes for obtaining prognostic metrics in vivo. [165]. Oxygen saturation measured by BOLD fMRI may serve to illuminate VCA reperfusion-injury to elucidate mechanisms of graft deterioration. Expanding upon this method, four-dimensional flow MRI increases data acquisition to derive hemodynamic parameters such as shear stress, pressure change, turbulent kinetic energy, and pulse wave velocity [166]. This has been applied extensively in cardiology to obtain detailed images of blood flow through the cardiovascular system and may be adapted to assessment of VCA perfusion efficacy and vascular integrity [167]. However, the high resolution images obtained by fMRI necessitate powerful equipment. MRI machines of up to 7 Tesla were used in aforementioned hemodynamic studies. Currently, MRI miniaturization, while being aggressively pursued, has not reached the stage of portability needed for transplant monitoring [168]. Subsequently, MRI serves more effectively to assess VCA transplants in vivo for long-term study of vasculature.

Ultrasound imaging

The most significant advantages of ultrasound over MRI are the widespread access to ultrasound machines and feasibility of real-time tissue assessment. In one instance, ultrasound biomicroscopy identified markers of chronic rejection in in vivo VCA by detecting transplant arteriopathy in face transplants [169]. The high resolution of ultrasound biomicroscopy enables measurement of blood vessel wall thickening, currently explored as an indicator of graft vasculopathy and potential marker of chronic rejection [170, 171]. Ultrasound has also been used to intraoperatively assess blood flow patency on the day of surgery and one week post-operatively to ensure blood vessels have been successfully connected [172]. Measurements of blood flow and vessel structure were used to demonstrate that the microsurgery and perfusion with blood did not induce subsequent graft deterioration. The high-resolution imaging afforded by ultrasound biomicroscopy is a promising application to allograft monitoring and has potential to be leveraged for ex vivo applications, although suitability for the complex tissue architecture of VCA has not been established.

A limited number of novel developments strive to leverage ultrasound to quantify observations of edema and to measure the depth of pitting edema [173,174,175]. In a clinical study, the relationship between surface imprint depth, circumferential measurement, and tissue thickness revealed that depth of surface imprints correlate with subcutaneous tissue thickness by a coefficient of 0.736 [174]. In a similar manner, Pitre and colleagues considered the viscoelasticity of tissue as a measure of edema status and simplified hardware by designing a single-element ultrasound transducer [175]. Graphing tissue viscoelasticity across simulated edema conditions indicated depth dependency, therefore providing enhanced quantification of superficial edema. Ultrasound-based prognostic methods provide advantages of being widely available, nondestructive, portable, and simple to use (Table 2). However, ultrasound analysis requires expert knowledge of anatomical features and requires further analysis for quantification. Furthermore, efforts to correlate echogenicity with edema have been limited to superficial tissues, emphasizing that sufficient resolution in deeper tissue has not been established. While moving toward clinically accessible technology, the feasibility of ultrasound in ex vivo transplant medicine is currently limited by the lack of robust quantification and hardware compaction yet has promise as a non-destructive and high-resolution tissue monitoring tool.

Bioimpedance in monitoring and imaging

Bioimpedance spectroscopy (BIS) and electrical impedance tomography (EIT) are methods entrenched in diverse fields including agriculture, nutrition, and cellular biology [176,177,178]. To measure bioimpedance, an electric current is injected through the tissue then potential difference is collected [99]. Bioimpedance is calculated as the ratio of the injected current and collected potential. By varying the frequency of the current, specific cellular compartments can be probed. At lower frequencies, the current flows through the extracellular space. As frequency increases, current passes through the cell membrane and into the intracellular space [179]. Properties such as tissue density, cellular integrity, and fluid accumulation contribute to the total impedance. The application of bioimpedance for edema detection is a well-established method in lymphedema diagnosis [180]. Additionally, bioimpedance sensing has been used to monitor edema manifesting in cerebral hemorrhage, colitis, and in superficial tissue [148, 181,182,183,184,185,186].

The current range of noninvasive methods to evaluate VCA includes high-contrast, spatiotemporal imaging sufficient to resolve relevant pathophysiological tissue features such as vasculopathy and interstitial edema. However, these methods often require sophisticated instrumentation and specialized equipment. Bioimpedance analysis uses relatively minimal instrumentation for quantitative tissue assessment based on intrinsic electrical properties, with subcellular and tissue resolution depending on applied frequency ranges [187,188,189,190,191,192,193]. Limitations to bioimpedance applications in VCA include the lack of standardization in protocols, as well as unknown baselines for parameters measured. Standardization including optimal electrode placement, frequencies for measuring VCA impedance, and magnitude and phase threshold of impedance values in varying tissue types must be established for robust tissue analysis [194,195,196]. Furthermore, the complex architecture of VCA complicates analysis since the flow of electric current and therefore impedance depends on the orientation of cells [197]. Additionally, impedance measurements are affected by the anisotropic properties of nerve fibers, muscle, and blood vessels [198]. Judicious electrode configurations that mitigate anisotropic effects should be considered when applying impedance to tissue analysis [199]. The prospect of multimodal data fusion (MMDF) of temporal bioimpedance data, reflective of edema, with metabolic and cytokine profile data, and histology data to achieve VCA stratification has recently been advanced [100].

Opportunities and future directions

While much work is still necessary for the clinical acceptance of methods other than static cold storage, evidence for effective and novel VCA preservation solutions shows promise. Introducing nutrient flow and metabolite removal may overcome consequences of ischemia. Maintaining normothermia limits tissue damage associated with cold storage. Long-term VCA outcomes stand to be improved by developments in perfusion technology including electrical myostimulation (EMS) to maintain muscle tone and reduce atrophy [204, 205] support endothelial angiogenesis [206], and support nerve regeneration [207,208,209]. Before these methods can be established, it is necessary to verify improvements in tissue status to evaluate the merits of novel preservation solutions. Decline in tissue viability occurs progressively during storage and is associated with molecular and physical biomarkers. Current preservation strategies strive to prolong allograft viability and mitigate rejection by reducing pathophysiologic symptoms. Ex vivo tissue analysis can provide an empirical basis for clinical decisions to discard, treat, or transplant allografts. Ultimately, robust diagnostic monitoring can complement therapeutics to expand VCA feasibility.

Technological advancement

Given the relatively recent development of VCA surgery, progress in the diversity of tissue assessment modalities has been relatively slow, but promising. Liquid biopsies allow quantitative observation of cellular and molecular tissue changes. Noninvasive tools from solid organ transplantation have been adapted to visualize tissues of VCA on a microvascular scale. While insightful, further technological advancement is necessary for use in ex vivo tissue monitoring. Rather than using sample tissue or perfusate for proteomic and metabolomic analysis, in-line sensors complement machine perfusion to enable real-time and continuous monitoring to enable automated adaptation using perfusate supplements [70]. To meet the needs of VCA, commercial in-line sensors must be calibrated to specific models of ex vivo grafts and their perfusion conditions; moreover further improvement in their limit of detection is required to permit detection on a sub-millimolar scale. The non-invasive tools that were discussed in this review rely principally on conventional medical imaging technologies. A significant area of development for ex vivo monitoring is hardware size, as many high resolution methods lack portability. While recent technological advancements have enabled miniaturization, image analysis remains semi-quantitative and requires substantial expertise for data analysis and interpretation. A promising avenue of development leverages bioimpedance for non-invasive and continuous tissue evaluation. However, further investigation is warranted as the electrical properties of tissues are impacted by tissue composition, fluid retention, differences in preservation conditions, and other experimental variables [210].

Graft pre-conditioning

As a step beyond VCA preservation, recent developments in machine perfusion apply graft conditioning prior to transplantation. Both immunomodulatory molecules and hematopoietic cells have been used to lower the probability of transplant rejection [18, 57, 211,212,213,214,215,216]. As discussed, VCA is highly immunogenic and presents a high risk of acute rejection. While rejection can be treated following transplantation with standard immunosuppression regimens, medication is also a significant source of non-compliance due to the high volume needed [217]. Notable instances of acute rejection have occurred in which grafts had to be removed due to non-compliance with medication [218,219,220]. Furthermore, long-term immunosuppression increases the risk of developing infectious complications. Immunomodulation of the VCA to immunologically condition the graft prior to transplantation may enhance graft acceptance by the recipient immune system and potentially reduce immunosuppressive treatment regimens.

Conclusion

Vascularized composite allotransplantation is a complex and multifaceted procedure that may significantly improve patient quality of life. However, logistical challenges and heightened risks of immune rejection limit the number of feasible procedures. These challenges are compounded by damage-susceptible muscle and nerve tissues, which must remain intact to restore functional motor capabilities. The development of effective preservation methods would allow the potential to perform VCA transplantation outside the restrictive 4- to 6-h time window of current standard methods, thus expanding the pool of acceptable allografts and buying time for complex transplantation procedures without compromising graft functionality. As further research seeks to develop methods to minimize graft damage while sustaining viability for extended periods of time, advancements in status monitoring technology must establish their efficacy. Quantitative, substantive evidence for the efficacy of ex vivo perfusion and varying storage conditions are currently limited. To bridge the gap between modern technology and VCA transplantation, it is necessary to develop analytical tools that evaluate surrogate biomarkers of tissue status and connect their significance to transplant outcomes.

References

  1. Kollar B, Pomahac B, Riella LV. Novel immunological and clinical insights in vascularized composite allotransplantation. Curr Opin Organ Transplant. 2019;24(1):42–8. https://doi.org/10.1097/MOT.0000000000000592.

    Article  CAS  PubMed  Google Scholar 

  2. Haug V, Kollar B, Obed D, et al. The evolving clinical presentation of acute rejection in facial transplantation. JAMA Facial Plast Surg. 2019;21(4):278–85. https://doi.org/10.1001/jamafacial.2019.0076.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Shores JT, Malek V, Lee WPA, Brandacher G. Outcomes after hand and upper extremity transplantation. J Mater Sci Mater Med. 2017;28(5):72. https://doi.org/10.1007/s10856-017-5880-0.

    Article  CAS  PubMed  Google Scholar 

  4. Diaz-Siso JR, Parker M, Bueno EM, et al. Facial allotransplantation: a 3-year follow-up report. J Plast Reconstr Aesthetic Surg JPRAS. 2013;66(11):1458–63. https://doi.org/10.1016/j.bjps.2013.06.046.

    Article  PubMed  Google Scholar 

  5. Ramly EP, Kantar RS, Diaz-Siso JR, Alfonso AR, Shetye PR, Rodriguez ED. Outcomes after tooth-bearing maxillomandibular facial transplantation: insights and lessons learned. J Oral Maxillofac Surg. 2019;77(10):2085–103. https://doi.org/10.1016/j.joms.2019.05.011.

    Article  PubMed  Google Scholar 

  6. Singh M, Sisk G, Carty M, et al. Functional outcomes after bilateral hand transplantation: a 3.5-year comprehensive follow-up. Plast Reconstr Surg. 2016;137(1):185–9. https://doi.org/10.1097/PRS.0000000000001872.

    Article  CAS  PubMed  Google Scholar 

  7. Schneeberger S, Petruzzo P, Morelon E, et al. 20-Year follow-up of two cases of bilateral hand transplantation. N Engl J Med. 2020;383(18):1791–2. https://doi.org/10.1056/NEJMc2017711.

    Article  PubMed  Google Scholar 

  8. Ziegler-Graham K, MacKenzie EJ, Ephraim PL, Travison TG, Brookmeyer R. Estimating the prevalence of limb loss in the United States: 2005 to 2050. Arch Phys Med Rehabil. 2008;89(3):422–9. https://doi.org/10.1016/j.apmr.2007.11.005.

    Article  PubMed  Google Scholar 

  9. Biddiss EA, Chau TT. Upper limb prosthesis use and abandonment: a survey of the last 25 years. Prosthet Orthot Int. 2007;31(3):236–57. https://doi.org/10.1080/03093640600994581.

    Article  PubMed  Google Scholar 

  10. Chung KC, Oda T, Saddawi-Konefka D, Shauver MJ. An economic analysis of hand transplantation in the United States. Plast Reconstr Surg. 2010;125(2):589–98. https://doi.org/10.1097/PRS.0b013e3181c82eb6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Salminger S, Sturma A, Roche AD, et al. Functional and psychosocial outcomes of hand transplantation compared with prosthetic fitting in below-elbow amputees: a multicenter cohort study. PLoS ONE. 2016;11(9): e0162507. https://doi.org/10.1371/journal.pone.0162507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Messner F, Grahammer J, Hautz T, Brandacher G, Schneeberger S. Ischemia/reperfusion injury in vascularized tissue allotransplantation: tissue damage and clinical relevance. Curr Opin Organ Transplant. 2016;21(5):503–9. https://doi.org/10.1097/MOT.0000000000000343.

    Article  CAS  PubMed  Google Scholar 

  13. Wiedemann D, Schachner T, Bonaros N, et al. Impact of cold ischemia on mitochondrial function in porcine hearts and blood vessels. Int J Mol Sci. 2013;14(11):22042–51. https://doi.org/10.3390/ijms141122042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Brandão ML, Roselino JES, Piccinato CE, Cherri J. Mitochondrial alterations in skeletal muscle submitted to total ischemia. J Surg Res. 2003;110(1):235–40. https://doi.org/10.1016/s0022-4804(02)00093-8.

    Article  PubMed  Google Scholar 

  15. Herzberg G, Weppe F, Masson N, Gueffier X, Erhard L. Clinical evaluation of two bilateral hand allotransplantations at six and three years follow-up. Chir Main. 2008;27(2–3):109–17. https://doi.org/10.1016/j.main.2008.02.002.

    Article  CAS  PubMed  Google Scholar 

  16. Piza-Katzer H, Ninkovic M, Pechlaner S, Gabl M, Ninkovic M, Hussl H. Double hand transplantation: functional outcome after 18 months. J Hand Surg Edinb Scotl. 2002;27(4):385–90. https://doi.org/10.1054/jhsb.2002.0759.

    Article  CAS  Google Scholar 

  17. Kueckelhaus M, Dermietzel A, Alhefzi M, et al. Acellular hypothermic extracorporeal perfusion extends allowable ischemia time in a porcine whole limb replantation model. Plast Reconstr Surg. 2017;139(4):922e–32e. https://doi.org/10.1097/PRS.0000000000003208.

    Article  CAS  PubMed  Google Scholar 

  18. Burlage LC, Tessier SN, Etra JW, Uygun K, Brandacher G. Advances in machine perfusion, organ preservation, and cryobiology: potential impact on VCA. Curr Opin Organ Transplant. 2018;23(5):561–7. https://doi.org/10.1097/MOT.0000000000000567.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Mendenhall SD, Ginnetti MT, Sawyer JD, et al. Prevalence and distribution of potential vascularized composite allotransplant donors, implications for optimizing the donor-recipient match. Plast Reconstr Surg Glob Open. 2018. https://doi.org/10.1097/GOX.0000000000001833.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Amin KR, Stone JP, Kerr J, et al. Randomized preclinical study of machine perfusion in vascularized composite allografts. BJS Br J Surg. 2020. https://doi.org/10.1002/bjs.11921.

    Article  Google Scholar 

  21. Datta N, Devaney SG, Busuttil RW, Azari K, Kupiec-Weglinski JW. Prolonged cold ischemia time results in local and remote organ dysfunction in a murine model of vascularized composite transplantation. Am J Transplant. 2017;17(10):2572–9. https://doi.org/10.1111/ajt.14290.

    Article  CAS  PubMed  Google Scholar 

  22. Gok E, Kubiak CA, Guy E, Kemp SWP, Ozer K. Effect of static cold storage on skeletal muscle after vascularized composite tissue allotransplantation. J Reconstr Microsurg. 2020;36(1):9–15. https://doi.org/10.1055/s-0039-1693455.

    Article  PubMed  Google Scholar 

  23. Walker PM. Ischemia/reperfusion injury in skeletal muscle. Ann Vasc Surg. 1991;5(4):399–402. https://doi.org/10.1007/BF02015307.

    Article  CAS  PubMed  Google Scholar 

  24. Gaudet AD, Popovich PG, Ramer MS. Wallerian degeneration: gaining perspective on inflammatory events after peripheral nerve injury. J Neuroinflamm. 2011;8:110. https://doi.org/10.1186/1742-2094-8-110.

    Article  Google Scholar 

  25. Zhou T, Prather ER, Garrison DE, Zuo L. Interplay between ROS and antioxidants during ischemia-reperfusion injuries in cardiac and skeletal muscle. Int J Mol Sci. 2018;19(2):417. https://doi.org/10.3390/ijms19020417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Müller S, Constantinescu MA, Kiermeir DM, et al. Ischemia/reperfusion injury of porcine limbs after extracorporeal perfusion. J Surg Res. 2013;181(1):170–82. https://doi.org/10.1016/j.jss.2012.05.088.

    Article  PubMed  Google Scholar 

  27. Caterson EJ, Lopez J, Medina M, Pomahac B, Tullius SG. Ischemia-reperfusion injury in vascularized composite allotransplantation. J Craniofac Surg. 2013;24(1):51–6. https://doi.org/10.1097/SCS.0b013e31827104e1.

    Article  PubMed  Google Scholar 

  28. Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012;298:229–317. https://doi.org/10.1016/B978-0-12-394309-5.00006-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Friedman O, Carmel N, Sela M, et al. Immunological and inflammatory mapping of vascularized composite allograft rejection processes in a rat model. PLoS One San Franc. 2017;12(7): e0181507. https://doi.org/10.1371/journal.pone.0181507.

    Article  CAS  Google Scholar 

  30. Datta S, Fitzpatrick AM, Haykal S. Preservation solutions for attenuation of ischemia–reperfusion injury in vascularized composite allotransplantation. SAGE Open Med. 2021;9:20503121211034924. https://doi.org/10.1177/20503121211034924.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Fuller B, Froghi F, Davidson B. Organ preservation solutions: linking pharmacology to survival for the donor organ pathway. Curr Opin Organ Transplant. 2018;23(3):361–8. https://doi.org/10.1097/MOT.0000000000000525.

    Article  CAS  PubMed  Google Scholar 

  32. Jing L, Yao L, Zhao M, Peng L-P, Liu M. Organ preservation: from the past to the future. Acta Pharmacol Sin. 2018;39(5):845–57. https://doi.org/10.1038/aps.2017.182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Robbins N, Wordsworth MJ, Sippel MR, et al. Prevention of ischemia-reperfusion injury and chronic rejection in a porcine vascularized composite allotransplantation model. Transplantation. 2018;102:S706. https://doi.org/10.1097/01.tp.0000543670.93080.cc.

    Article  Google Scholar 

  34. Ozer K, Rojas-Pena A, Mendias CL, Bryner BS, Toomasian C, Bartlett RH. The effect of ex situ perfusion in a swine limb vascularized composite tissue allograft on survival up to 24 hours. J Hand Surg. 2016;41(1):3–12. https://doi.org/10.1016/j.jhsa.2015.11.003.

    Article  Google Scholar 

  35. Belzer FO, Ashby BS, Huang JS, Dunphy JE. Etiology of rising perfusion pressure in isolated organ perfusion. Ann Surg. 1968;168(3):382–91. https://doi.org/10.1097/00000658-196809000-00008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Salehi S, Tran K, Grayson WL. Advances in perfusion systems for solid organ preservation. Yale J Biol Med. 2018;91(3):301–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hicks TE, Boswick JA, Solomons CC. The effects of perfusion on an amputated extremity. J Trauma. 1980;20(8):632–48.

    Article  CAS  PubMed  Google Scholar 

  38. Gordon L, Levinsohn DG, Borowsky CD, et al. Improved preservation of skeletal muscle in amputated limbs using pulsatile hypothermic perfusion with University of Wisconsin solution. A preliminary study. J Bone Jt Surg Ser A. 1992;74:1358–66. https://doi.org/10.2106/00004623-199274090-00009.

    Article  CAS  Google Scholar 

  39. Domingo-Pech J, Garriga JM, Toran N, et al. Preservation of the amputated canine hind limb by extracorporeal perfusion. Int Orthop. 1991;15(4):289–91. https://doi.org/10.1007/BF00186863.

    Article  CAS  PubMed  Google Scholar 

  40. Lapchinsky AG. Recent results of experimental transplantation of preserved limbs and kidneys and possible use of this technique in clinical practice. Ann N Y Acad Sci. 2006;87(1):539–71. https://doi.org/10.1111/j.1749-6632.1960.tb23220.x.

    Article  Google Scholar 

  41. Constantinescu MA, Knall E, Xu X, et al. Preservation of amputated extremities by extracorporeal blood perfusion; a feasibility study in a porcine model. J Surg Res. 2011;171(1):291–9. https://doi.org/10.1016/j.jss.2010.01.040.

    Article  PubMed  Google Scholar 

  42. Kruit AS, Smits L, Pouwels A, Schreinemachers MCJM, Hummelink SLM, Ulrich DJO. Ex-vivo perfusion as a successful strategy for reduction of ischemia-reperfusion injury in prolonged muscle flap preservation—a gene expression study. Gene. 2019;701:89–97. https://doi.org/10.1016/j.gene.2019.03.021.

    Article  CAS  PubMed  Google Scholar 

  43. Gok E, Alghanem F, Moon R, et al. Development of an ex-situ limb perfusion system for a rodent model. ASAIO J Am Soc Artif Intern Organs 1992. 2019;65(2):167–72. https://doi.org/10.1097/MAT.0000000000000786.

    Article  Google Scholar 

  44. Ozturk MB, Aksan T, Ozcelik IB, et al. Extracorporeal free flap perfusion using extracorporeal membrane oxygenation device: an experimental model. Ann Plast Surg. 2019;83(6):702–8. https://doi.org/10.1097/SAP.0000000000002014.

    Article  CAS  PubMed  Google Scholar 

  45. Kueckelhaus M, Fischer S, Sisk G, et al. A mobile extracorporeal extremity salvage system for replantation and transplantation. Ann Plast Surg. 2016;76(3):355–60. https://doi.org/10.1097/SAP.0000000000000681.

    Article  CAS  PubMed  Google Scholar 

  46. Cooley B, Tadych K, Gould J. Perfusion of free flaps with heparinized whole blood during ischemic storage. J Reconstr Microsurg. 1990;6(01):49–53. https://doi.org/10.1055/s-2007-1006802.

    Article  CAS  PubMed  Google Scholar 

  47. Werner NL, Alghanem F, Rakestraw SL, et al. Ex situ perfusion of human limb allografts for 24 hours. Transplantation. 2017;101(3):e68–74. https://doi.org/10.1097/TP.0000000000001500.

    Article  PubMed  Google Scholar 

  48. Lanzetta M, Petruzzo P, Margreiter R, et al. The international registry on hand and composite tissue transplantation. Transplantation. 2005;79(9):1210–4. https://doi.org/10.1097/01.TP.0000157118.28394.FA.

    Article  PubMed  Google Scholar 

  49. Menezes AQ, Pêgo-Fernandes PM, Cardoso PFG, et al. Comparison of Celsior and Perfadex lung preservation solutions in rat lungs subjected to 6 and 12 hours of ischemia using an ex-vivo lung perfusion system. Clin Sao Paulo Braz. 2012;67(11):1309–14. https://doi.org/10.6061/clinics/2012(11)15.

    Article  Google Scholar 

  50. Urcuyo D, Blum MF, Liu Q, et al. Development of a prolonged warm ex vivo perfusion model for kidneys donated after cardiac death. Int J Artif Organs. 2017;40(6):265–71. https://doi.org/10.5301/ijao.5000586.

    Article  PubMed  Google Scholar 

  51. Cobert ML, Peltz M, West LM, Jessen ME. Importance of organ preservation solution composition in reducing myocardial edema during machine perfusion for heart transplantation. Transplant Proc. 2010;42(5):1591–4. https://doi.org/10.1016/j.transproceed.2010.02.073.

    Article  CAS  PubMed  Google Scholar 

  52. Messner F, Hautz T, Blumer MJF, et al. Critical ischemia times and the effect of novel preservation solutions HTK-N and TiProtec on tissues of a vascularized tissue isograft. Transplantation. 2017;101(9): e301. https://doi.org/10.1097/TP.0000000000001845.

    Article  CAS  PubMed  Google Scholar 

  53. Haug V, Kollar B, Endo Y, et al. Comparison of acellular solutions for ex-situ perfusion of amputated limbs. Mil Med. 2020;185(11–12):e2004–12. https://doi.org/10.1093/milmed/usaa160.

    Article  PubMed  Google Scholar 

  54. Kruit AS, Brouwers K, van Midden D, et al. Successful 18-h acellular extracorporeal perfusion and replantation of porcine limbs—histology versus nerve stimulation. Transpl Int. 2021;34(2):365–75. https://doi.org/10.1111/tri.13802.

    Article  CAS  PubMed  Google Scholar 

  55. Etra JW, Grzelak MJ, Fidder SAJ, et al. A skin rejection grading system for vascularized composite allotransplantation in a preclinical large animal model. Transplantation. 2019;103(7):1385–91. https://doi.org/10.1097/TP.0000000000002695.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Cendales LC, Kanitakis J, Schneeberger S, et al. The Banff 2007 working classification of skin-containing composite tissue allograft pathology. Am J Transplant. 2008;8(7):1396–400. https://doi.org/10.1111/j.1600-6143.2008.02243.x.

    Article  CAS  PubMed  Google Scholar 

  57. Rosales IA, Foreman RK, DeFazio M, et al. Systematic pathological component scores for skin-containing vascularized composite allografts. Vasc Compos Allotransplant. 2016;3(1–2):62–74. https://doi.org/10.1080/23723505.2017.1318200.

    Article  Google Scholar 

  58. Kaufman CL, Marvin MR, Chilton PM, et al. Immunobiology in VCA. Transpl Int. 2016;29(6):644–54. https://doi.org/10.1111/tri.12764.

    Article  PubMed  Google Scholar 

  59. Kollar B, Uffing A, Borges TJ, et al. MMP3 is a non-invasive biomarker of rejection in skin-bearing vascularized composite allotransplantation: a multicenter validation study. Front Immunol. 2019. https://doi.org/10.3389/fimmu.2019.02771.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Chhablani J, Deepa MJ, Tyagi M, Narayanan R, Kozak I. Fluorescein angiography and optical coherence tomography in myopic choroidal neovascularization. Eye. 2015;29(4):519–24. https://doi.org/10.1038/eye.2014.345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Samper IC, Gowers SAN, Booth MA, et al. Portable microfluidic biosensing system for real-time analysis of microdialysate in transplant kidneys. Anal Chem. 2019;91(22):14631–8. https://doi.org/10.1021/acs.analchem.9b03774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kuzon WM, Walker PM, Mickle DA, Harris KA, Pynn BR, McKee NH. An isolated skeletal muscle model suitable for acute ischemia studies. J Surg Res. 1986;41(1):24–32. https://doi.org/10.1016/0022-4804(86)90004-1.

    Article  PubMed  Google Scholar 

  63. Obara H, Matsubara K, Kitagawa Y. Acute limb ischemia. Ann Vasc Dis. 2018;11(4):443–8. https://doi.org/10.3400/avd.ra.18-00074.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Couto-Mallón D, González-Vílchez F, Almenar-Bonet L, et al. Prognostic value of serum lactate levels in patients undergoing urgent heart transplant: a subanalysis of the ASIS-TC Spanish multicenter study. Rev Esp Cardiol Engl Ed. 2019;72(3):208–14. https://doi.org/10.1016/j.rec.2018.02.021.

    Article  PubMed  Google Scholar 

  65. Wu JF, Wu RY, Chen J, Ou-Yang B, Chen MY, Guan XD. Early lactate clearance as a reliable predictor of initial poor graft function after orthotopic liver transplantation. Hepatobiliary Pancreat Dis Int HBPD Int. 2011;10(6):587–92. https://doi.org/10.1016/s1499-3872(11)60100-8.

    Article  CAS  PubMed  Google Scholar 

  66. Schraufnagel DP, Steffen RJ, Vargo PR, et al. Devices for ex vivo heart and lung perfusion. Expert Rev Med Devices. 2018;15(3):183–91. https://doi.org/10.1080/17434440.2018.1430568.

    Article  CAS  PubMed  Google Scholar 

  67. Benzon HT, Toleikis JR, Meagher LL, Shapiro BA, Ts’ao CH, Avram MJ. Changes in venous blood lactate, venous blood gases, and somatosensory evoked potentials after tourniquet application. Anesthesiology. 1988;69(5):677–82. https://doi.org/10.1097/00000542-198811000-00007.

    Article  CAS  PubMed  Google Scholar 

  68. Fahradyan V, Said SA, Ordenana C, et al. Extended ex vivo normothermic perfusion for preservation of vascularized composite allografts. Artif Organs. 2020;44(8):846–55. https://doi.org/10.1111/aor.13678.

    Article  CAS  PubMed  Google Scholar 

  69. Gok E, Kubiak CA, Guy E, et al. Long-term effects of hypothermic ex situ perfusion on skeletal muscle metabolism, structure, and force generation after transplantation. Transplantation. 2019;103(10):2105–12. https://doi.org/10.1097/TP.0000000000002800.

    Article  CAS  PubMed  Google Scholar 

  70. Eshmuminov D, Becker D, Bautista Borrego L, et al. An integrated perfusion machine preserves injured human livers for 1 week. Nat Biotechnol. 2020;38(2):189–98. https://doi.org/10.1038/s41587-019-0374-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Garcia-Dorado D, Ruiz-Meana M, Inserte J, Rodriguez-Sinovas A, Piper HM. Calcium-mediated cell death during myocardial reperfusion. Cardiovasc Res. 2012;94(2):168–80. https://doi.org/10.1093/cvr/cvs116.

    Article  CAS  PubMed  Google Scholar 

  72. Paradis S, Charles AL, Meyer A, et al. Chronology of mitochondrial and cellular events during skeletal muscle ischemia-reperfusion. Am J Physiol Cell Physiol. 2016;310(11):C968–82. https://doi.org/10.1152/ajpcell.00356.2015.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Hausenloy DJ, Yellon DM. The mitochondrial permeability transition pore: its fundamental role in mediating cell death during ischaemia and reperfusion. J Mol Cell Cardiol. 2003;35(4):339–41. https://doi.org/10.1016/S0022-2828(03)00043-9.

    Article  CAS  PubMed  Google Scholar 

  74. Borutaite V, Jekabsone A, Morkuniene R, Brown GC. Inhibition of mitochondrial permeability transition prevents mitochondrial dysfunction, cytochrome c release and apoptosis induced by heart ischemia. J Mol Cell Cardiol. 2003;35(4):357–66. https://doi.org/10.1016/S0022-2828(03)00005-1.

    Article  CAS  PubMed  Google Scholar 

  75. Ivanics T, Miklós Z, Ruttner Z, et al. Ischemia/reperfusion-induced changes in intracellular free Ca2+ levels in rat skeletal muscle fibers—an in vivo study. Pflugers Arch. 2000;440(2):302–8. https://doi.org/10.1007/s004240000287.

    Article  CAS  PubMed  Google Scholar 

  76. Akande O, Chen Q, Toldo S, Lesnefsky EJ, Quader M. Ischemia and reperfusion injury to mitochondria and cardiac function in donation after circulatory death hearts—an experimental study. PLoS ONE. 2020;15(12): e0243504. https://doi.org/10.1371/journal.pone.0243504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kruit AS, Schreinemachers MCJM, Koers EJ, Zegers HJH, Hummelink S, Ulrich DJO. Successful long-term extracorporeal perfusion of free musculocutaneous flaps in a porcine model. J Surg Res. 2019;235:113–23. https://doi.org/10.1016/j.jss.2018.09.076.

    Article  PubMed  Google Scholar 

  78. Rahman ARA, Justin G, Guiseppi-Wilson A, Guiseppi-Elie A. Fabrication and packaging of a dual sensing electrochemical biotransducer for glucose and lactate useful in intramuscular physiologic status monitoring. IEEE Sens J. 2009;9(12):1856–63. https://doi.org/10.1109/JSEN.2009.2031347.

    Article  CAS  Google Scholar 

  79. Kotanen C, Guiseppi-Elie A. Development of an implantable biosensor system for physiological status monitoring during long duration space exploration. Gravit Space Biol. 2010;23(2):55–64.

    Google Scholar 

  80. Guiseppi-Elie A. An implantable biochip to influence patient outcomes following trauma-induced hemorrhage. Anal Bioanal Chem. 2011;399(1):403–19. https://doi.org/10.1007/s00216-010-4271-x.

    Article  CAS  PubMed  Google Scholar 

  81. Carrara S, Ghoreishizadeh S, Olivo J, et al. Fully integrated biochip platforms for advanced healthcare. Sensors. 2012;12(8):11013–60. https://doi.org/10.3390/s120811013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kotanen CN, Guiseppi-Elie A. Monitoring systems and quantitative measurement of biomolecules for the management of trauma. Biomed Microdevices. 2013;15(3):561–77. https://doi.org/10.1007/s10544-013-9756-x.

    Article  CAS  PubMed  Google Scholar 

  83. Aggas JR, Sánchez-Sinencio E, Guiseppi-Elie A. Wien oscillator using organic enzyme-chemiresistors for fused measurement of glucose and lactate. Adv Intell Syst. 2020;2(7):2000004. https://doi.org/10.1002/aisy.202000004.

    Article  Google Scholar 

  84. Bhat A, Podstawczyk D, Walther BK, et al. Toward a hemorrhagic trauma severity score: fusing five physiological biomarkers. J Transl Med. 2020;18(1):348. https://doi.org/10.1186/s12967-020-02516-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gok MA, Buckley PE, Shenton BK, et al. Long-term renal function in kidneys from non-heart-beating donors: a single-center experience. Transplantation. 2002;74(5):664–9.

    Article  PubMed  Google Scholar 

  86. Ozer K, Rojas-Pena A, Mendias CL, Bryner B, Toomasian C, Bartlett RH. Ex situ limb perfusion system to extend vascularized composite tissue allograft survival in swine. Transplantation. 2015;99(10):2095–101. https://doi.org/10.1097/TP.0000000000000756.

    Article  PubMed  Google Scholar 

  87. Lledo-Garcia E, Hernandez-Fernandez C, Pedemonte G, Rodriguez-Martinez D, Tejedor-Jorge A, del Cañizo-Lopez JF. Effects of hypothermic perfusion in isolated pig kidneys after warm ischemia. Transplant Proc. 2007;39(1):258–60. https://doi.org/10.1016/j.transproceed.2006.10.197.

    Article  CAS  PubMed  Google Scholar 

  88. Orloff MS, Reed AI, Erturk E, et al. Nonheartbeating cadaveric organ donation. Ann Surg. 1994;220(4):578–83; discussion 583-585. https://doi.org/10.1097/00000658-199410000-00016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Jochmans I, Moers C, Smits JM, et al. The prognostic value of renal resistance during hypothermic machine perfusion of deceased donor kidneys. Am J Transplant. 2011;11(10):2214–20. https://doi.org/10.1111/j.1600-6143.2011.03685.x.

    Article  CAS  PubMed  Google Scholar 

  90. Brook NR, Knight AJ, Nicholson ML. Intra-renal resistance reflects warm ischaemic damage, and is further increased by static cold storage: a model of non-heart-beating donor kidneys. Med Sci Monit Int Med J Exp Clin Res. 2003;9(7):BR271-275.

    Google Scholar 

  91. Cheung Y fai. CHAPTER 6 - Systemic Circulation. In: Anderson RH, Baker EJ, Penny DJ, et al. editors. Paediatric cardiology (Third Edition). Churchill Livingstone; 2010. pp 91–116. https://doi.org/10.1016/B978-0-7020-3064-2.00006-0

  92. Walther BK, Pandian NKR, Gold KA, et al. Mechanotransduction-on-chip: vessel-chip model of endothelial YAP mechanobiology reveals matrix stiffness impedes shear response. Lab Chip. 2021;21(9):1738–51. https://doi.org/10.1039/d0lc01283a.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hosgood SA, Nicholson ML. First in man renal transplantation after ex vivo normothermic perfusion. Transplantation. 2011;92(7):735–8. https://doi.org/10.1097/TP.0b013e31822d4e04.

    Article  PubMed  Google Scholar 

  94. Minor T, Sitzia M, Dombrowski F. Kidney transplantation from non-heart-beating donors after oxygenated low-flow machine perfusion preservation with histidine-tryptophan-ketoglutarate solution. Transpl Int. 2005;17(11):707–12. https://doi.org/10.1007/s00147-004-0795-3.

    Article  CAS  PubMed  Google Scholar 

  95. Post ICJH, Dirkes MC, Heger M, Bezemer R, van’t Leven J, van Gulik TM. Optimal flow and pressure management in machine perfusion systems for organ preservation. Ann Biomed Eng. 2012;40(12):2698–707. https://doi.org/10.1007/s10439-012-0601-9.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Tsuji N, Yamashita S, Sugawara Y, Kobayashi E. Effect of prolonged ischaemic time on muscular atrophy and regenerating nerve fibres in transplantation of the rat hind limb. J Plast Surg Hand Surg. 2012;46(3–4):217–21. https://doi.org/10.3109/2000656X.2012.709726.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Taeger CD, Friedrich O, Dragu A, et al. Assessing viability of extracorporeal preserved muscle transplants using external field stimulation: a novel tool to improve methods prolonging bridge-to-transplantation time. Sci Rep. 2015;5(1):11956. https://doi.org/10.1038/srep11956.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Abasi S, Aggas JR, Venkatesh N, Vallavanatt IG, Guiseppi-Elie A. Design, fabrication and testing of an electrical cell stimulation and recording apparatus (ECSARA) for cells in electroculture. Biosens Bioelectron. 2020;147: 111793. https://doi.org/10.1016/j.bios.2019.111793.

    Article  CAS  PubMed  Google Scholar 

  99. Abasi S, Aggas JR, Garayar-Leyva GG, Walther BK, Guiseppi-Elie A. Bioelectrical impedance spectroscopy for monitoring mammalian cells and tissues under different frequency domains: a review. ACS Meas Sci Au. 2022;2(6):495–516. https://doi.org/10.1021/acsmeasuresciau.2c00033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Aggas JR, Abasi S, Ton C, et al. Real-time monitoring using multiplexed multi-electrode bioelectrical impedance spectroscopy for the stratification of vascularized composite allografts: a perspective on predictive analytics. Bioengineering. 2023;10(4):434. https://doi.org/10.3390/bioengineering10040434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Abasi S, Jain A, Cooke JP, Guiseppi-Elie A. Electrically stimulated gene expression under exogenously applied electric fields. Front Mol Biosci. 2023;10:1161191. https://doi.org/10.3389/fmolb.2023.1161191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kolegraff K, Kaufman C, Brandacher G. Defining rejection in vascularized composite allotransplantation: more than just arguing semantics. Vasc Compos Allotransplant. 2015;2(4):65–70. https://doi.org/10.1080/23723505.2016.1229650.

    Article  Google Scholar 

  103. Sarhane KA, Tuffaha SH, Broyles JM, et al. A critical analysis of rejection in vascularized composite allotransplantation: clinical, cellular and molecular aspects, current challenges, and novel concepts. Front Immunol. 2013. https://doi.org/10.3389/fimmu.2013.00406.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Shimizu F, Okamoto O, Katagiri K, Fujiwara S, Wei FC. Prolonged ischemia increases severity of rejection in skin flap allotransplantation in rats. Microsurgery. 2010;30(2):132–7. https://doi.org/10.1002/micr.20728.

    Article  PubMed  Google Scholar 

  105. Baumeister SP, Ofer N, Kleist C, et al. Comparison of six methods for the assessment of ischemia-reperfusion injury in skeletal muscle following composite tissue allotransplantation. J Reconstr Microsurg. 2004;20(03):253–9. https://doi.org/10.1055/s-2004-823113.

    Article  PubMed  Google Scholar 

  106. Sosin M, Mundinger GS, Drachenberg CB, Rodriguez ED. Lymphatic reconstitution and regeneration after face transplantation. Ann Plast Surg. 2017;79(5):505–8. https://doi.org/10.1097/SAP.0000000000001222.

    Article  CAS  PubMed  Google Scholar 

  107. Hautz T, Zelger BG, Weißenbacher A, et al. Standardizing skin biopsy sampling to assess rejection in vascularized composite allotransplantation. Clin Transplant. 2013;27(2):E81–90. https://doi.org/10.1111/ctr.12086.

    Article  PubMed  Google Scholar 

  108. Starzl R, Brandacher G, Lee WPA, et al. Review of the early diagnoses and assessment of rejection in vascularized composite allotransplantation. Clin Dev Immunol. 2013;2013: e402980. https://doi.org/10.1155/2013/402980.

    Article  CAS  Google Scholar 

  109. Eikmans M, Gielis EM, Ledeganck KJ, Yang J, Abramowicz D, Claas FFJ. Non-invasive biomarkers of acute rejection in kidney transplantation: novel targets and strategies. Front Med. 2019;5:358. https://doi.org/10.3389/fmed.2018.00358.

    Article  Google Scholar 

  110. Altara R, Manca M, Hessel MH, et al. CXCL10 is a circulating inflammatory marker in patients with advanced heart failure: a pilot study. J Cardiovasc Transl Res. 2016;9(4):302–14. https://doi.org/10.1007/s12265-016-9703-3.

    Article  PubMed  Google Scholar 

  111. Nankivell BJ, Alexander SI. Rejection of the kidney allograft. N Engl J Med. 2010;363(15):1451–62. https://doi.org/10.1056/NEJMra0902927.

    Article  CAS  PubMed  Google Scholar 

  112. Chin C, Chen G, Sequeria F, et al. Clinical usefulness of a novel C1q assay to detect immunoglobulin G antibodies capable of fixing complement in sensitized pediatric heart transplant patients. J Heart Lung Transplant. 2011;30(2):158–63. https://doi.org/10.1016/j.healun.2010.08.020.

    Article  PubMed  Google Scholar 

  113. Brancaccio P, Lippi G, Maffulli N. Biochemical markers of muscular damage. Clin Chem Lab Med. 2010;48(6):757–67. https://doi.org/10.1515/CCLM.2010.179.

    Article  CAS  PubMed  Google Scholar 

  114. Klocke FJ, Copley DP, Krawczyk JA, Reichlin M. Rapid renal clearance of immunoreactive canine plasma myoglobin. Circulation. 1982;65(7):1522–8. https://doi.org/10.1161/01.cir.65.7.1522.

    Article  CAS  PubMed  Google Scholar 

  115. Honeyman C, Stark H, Wang HC, Hester J, Issa F, Giele H. Biomarker and surrogate development in vascularised composite allograft transplantation: current progress and future challenges. J Plast Reconstr Aesthet Surg. 2020. https://doi.org/10.1016/j.bjps.2020.11.022.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Puscz F, Dadras M, Dermietzel A, et al. A chronic rejection model and potential biomarkers for vascularized composite allotransplantation. PLoS ONE. 2020;15(6): e0235266. https://doi.org/10.1371/journal.pone.0235266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Kollar B, Shubin A, Borges TJ, et al. Increased levels of circulating MMP3 correlate with severe rejection in face transplantation. Sci Rep. 2018;8(1):14915. https://doi.org/10.1038/s41598-018-33272-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. De Vlaminck I, Valantine HA, Snyder TM, et al. Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection. Sci Transl Med. 2014;6(241):241ra77. https://doi.org/10.1126/scitranslmed.3007803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Kueckelhaus M, Imanzadeh A, Fischer S, et al. Noninvasive monitoring of immune rejection in face transplant recipients. Plast Reconstr Surg. 2015;136(5):1082–9. https://doi.org/10.1097/PRS.0000000000001703.

    Article  CAS  PubMed  Google Scholar 

  120. Kataria A, Kumar D, Gupta G. Donor-derived cell-free DNA in solid-organ transplant diagnostics: indications, limitations, and future directions. Transplantation. 2021;105(6):1203–11. https://doi.org/10.1097/TP.0000000000003651.

    Article  CAS  PubMed  Google Scholar 

  121. Knight SR, Thorne A, Lo Faro ML. Donor-specific cell-free DNA as a biomarker in solid organ transplantation. A systematic review. Transplantation. 2019;103(2):273–83. https://doi.org/10.1097/TP.0000000000002482.

    Article  CAS  PubMed  Google Scholar 

  122. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–90. https://doi.org/10.1038/nm.1789.

    Article  CAS  PubMed  Google Scholar 

  123. McClure T, Goh SK, Cox D, Muralidharan V, Dobrovic A, Testro AG. Donor-specific cell-free DNA as a biomarker in liver transplantation: a review. World J Transplant. 2020;10(11):307–19. https://doi.org/10.5500/wjt.v10.i11.307.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Haug V, Kuang Y, Tasigiorgos S, et al. Circulating donor-derived cell-free DNA as a biomarker in vascularized composite allotransplantation? Transplantation. 2020. https://doi.org/10.1097/TP.0000000000003000.

    Article  PubMed  Google Scholar 

  125. Cretoiu (Ciontea) S. Circulating microRNAs in cardiovascular diseases: recent progress and challenges. J Hypertens Res. 2016;2:15–8.

    Google Scholar 

  126. Yokoi A, Matsuzaki J, Yamamoto Y, et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat Commun. 2018;9(1):4319. https://doi.org/10.1038/s41467-018-06434-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Paunescu IA, Bardan R, Marcu A, et al. Biomarker potential of plasma microRNA-150-5p in prostate cancer. Med Kaunas Lith. 2019;55(9):E564. https://doi.org/10.3390/medicina55090564.

    Article  Google Scholar 

  128. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99(24):15524–9. https://doi.org/10.1073/pnas.242606799.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–8. https://doi.org/10.1038/nature03702.

    Article  CAS  PubMed  Google Scholar 

  130. Di Stefano AB, Pappalardo M, Moschella F, Cordova A, Toia F. MicroRNAs in solid organ and vascularized composite allotransplantation: potential biomarkers for diagnosis and therapeutic use. Transplant Rev Orlando Fla. 2020;34(4): 100566. https://doi.org/10.1016/j.trre.2020.100566.

    Article  Google Scholar 

  131. Khalid U, Newbury LJ, Simpson K, et al. A urinary microRNA panel that is an early predictive biomarker of delayed graft function following kidney transplantation. Sci Rep. 2019;9(1):3584. https://doi.org/10.1038/s41598-019-38642-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Afshari A, Yaghobi R, Karimi MH, Mowla J. Alterations in microRNA gene expression profile in liver transplant patients with hepatocellular carcinoma. BMC Gastroenterol. 2021;21(1):262. https://doi.org/10.1186/s12876-020-01596-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Matton APM, Selten JW, Roest HP, et al. Cell-free microRNAs as early predictors of graft viability during ex vivo normothermic machine perfusion of human donor livers. Clin Transplant. 2020;34(3): e13790. https://doi.org/10.1111/ctr.13790.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Roth ES, Buck DG, Gorantla VS, Losee JE, Foust DE, Britton CA. The role of imaging in patient selection, preoperative planning, and postoperative monitoring in human upper extremity allotransplantation. J Transplant. 2014. https://doi.org/10.1155/2014/169546.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Weinzweig N, Gonzalez M. Free tissue failure is not an all-or-none phenomenon. Plast Reconstr Surg. 1995;96(3):648–60.

    Article  CAS  PubMed  Google Scholar 

  136. Kamphuis ME, Greuter MJW, Slart RHJA, Slump CH. Quantitative imaging: systematic review of perfusion/flow phantoms. Eur Radiol Exp. 2020;4(1):15. https://doi.org/10.1186/s41747-019-0133-2.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Gkontra P, El-Bouri WK, Norton KA, et al. Dynamic changes in microvascular flow conductivity and perfusion after myocardial infarction shown by image-based modeling. J Am Heart Assoc. 2019. https://doi.org/10.1161/JAHA.118.011058.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Hallock GG, Rice DC. A comparison of pulse oximetry and laser Doppler flowmetry in monitoring sequential vascular occlusion in a rabbit ear model. Can J Plast Surg. 2003;11(1):11–4.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Lese I, Leclère FM, Gayanayake T, et al. Regional lymphatic inclusion in orthotopic hindlimb transplantation: establishment and assessment of feasibility in a rodent model. Transplant Direct. 2020. https://doi.org/10.1097/TXD.0000000000001044.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Mücke T, Hapfelmeier A, Schmidt LH, et al. A comparative analysis using flowmeter, laser-Doppler spectrophotometry, and indocyanine green-videoangiography for detection of vascular stenosis in free flaps. Sci Rep. 2020;10(1):939. https://doi.org/10.1038/s41598-020-57777-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Bagavathiappan S, Saravanan T, Philip J, et al. Infrared thermal imaging for detection of peripheral vascular disorders. J Med Phys Assoc Med Phys India. 2009;34(1):43–7. https://doi.org/10.4103/0971-6203.48720.

    Article  CAS  Google Scholar 

  142. Ioannou S. Functional infrared thermal imaging: a contemporary tool in soft tissue screening. Sci Rep. 2020;10(1):9303. https://doi.org/10.1038/s41598-020-66397-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Lin PH, Saines M. Assessment of lower extremity ischemia using smartphone thermographic imaging. J Vasc Surg Cases Innov Tech. 2017;3(4):205–8. https://doi.org/10.1016/j.jvscit.2016.10.012.

    Article  PubMed  PubMed Central  Google Scholar 

  144. O’Donnell TF, Rasmussen JC, Sevick-Muraca EM. New diagnostic modalities in the evaluation of lymphedema. J Vasc Surg Venous Lymphat Disord. 2017;5(2):261–73. https://doi.org/10.1016/j.jvsv.2016.10.083.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Qin W, Wang RK. Assessment of edema volume in skin upon injury in a mouse ear model with optical coherence tomography. Lasers Med Sci. 2016;31(7):1351–61. https://doi.org/10.1007/s10103-016-1984-x.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Zaleska MT, Olszewski WL. Indocyanine green near-infrared lymphangiography for evaluation of effectiveness of edema fluid flow under therapeutic compression. J Biophotonics. 2018;11(8): e201700150. https://doi.org/10.1002/jbio.201700150.

    Article  CAS  PubMed  Google Scholar 

  147. Fichter AM, Ritschl LM, Borgmann A, et al. Development of an extracorporeal perfusion device for small animal free flaps. PLoS ONE. 2016;11(1): e0147755. https://doi.org/10.1371/journal.pone.0147755.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Cross KM, Leonardi L, Gomez M, et al. Noninvasive measurement of edema in partial thickness burn wounds. J Burn Care Res. 2009;30(5):807–17. https://doi.org/10.1097/BCR.0b013e3181b485e9.

    Article  PubMed  Google Scholar 

  149. Rasmussen JC, Tan IC, Marshall MV, Fife CE, Sevick-Muraca EM. Lymphatic imaging in humans with near-infrared fluorescence. Curr Opin Biotechnol. 2009;20(1):74–82. https://doi.org/10.1016/j.copbio.2009.01.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Hayn D, Fruhwald F, Riedel A, Falgenhauer M, Schreier G. Leg edema quantification for heart failure patients via 3D imaging. Sensors. 2013;13(8):10584–98. https://doi.org/10.3390/s130810584.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Badaró E, Novais E, Prodocimo LM, Sallum JMF. Spectral-domain optical coherence tomography for macular edema. Sci World J. 2014;2014: e191847. https://doi.org/10.1155/2014/191847.

    Article  Google Scholar 

  152. Trichonas G, Kaiser PK. Optical coherence tomography imaging of macular oedema. Br J Ophthalmol. 2014;98(Suppl 2):ii24–9. https://doi.org/10.1136/bjophthalmol-2014-305305.

    Article  PubMed  Google Scholar 

  153. Virgili G, Menchini F, Casazza G, et al. Optical coherence tomography (OCT) for detection of macular oedema in patients with diabetic retinopathy. Cochrane Database Syst Rev. 2015;1: CD008081. https://doi.org/10.1002/14651858.CD008081.pub3.

    Article  PubMed  Google Scholar 

  154. Chen H, Zhang Y, Pang J, et al. The differentiation of soft tissue infiltration and surrounding edema in an animal model of malignant bone tumor: evaluation by dual-energy CT. Technol Cancer Res Treat. 2019. https://doi.org/10.1177/1533033819846842.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Houben RH, Thaler R, Kotsougiani D, et al. Neo-angiogenesis, transplant viability, and molecular analyses of vascularized bone allotransplantation surgery in a large animal model. J Orthop Res. 2020;38(2):288–96. https://doi.org/10.1002/jor.24481.

    Article  CAS  PubMed  Google Scholar 

  156. Chen L, Ma H, Liu H, et al. Quantitative photoacoustic imaging for early detection of muscle ischemia injury. Am J Transl Res. 2017;9(5):2255–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Hayeri MR, Ziai P, Shehata ML, Teytelboym OM, Huang BK. Soft-tissue infections and their imaging mimics: from cellulitis to necrotizing fasciitis. Radiographics. 2016;36(6):1888–910. https://doi.org/10.1148/rg.2016160068.

    Article  PubMed  Google Scholar 

  158. McMahon CJ, Wu JS, Eisenberg RL. Muscle edema. Am J Roentgenol. 2010;194(4):W284–92. https://doi.org/10.2214/AJR.10.4247.

    Article  Google Scholar 

  159. Tada Y, Yang PC. Myocardial edema on T2-weighted MRI. Circ Res. 2017;121(4):326–8. https://doi.org/10.1161/CIRCRESAHA.117.311494.

    Article  CAS  PubMed  Google Scholar 

  160. Hueper K, Gutberlet M, Bräsen JH, et al. Multiparametric functional MRI: non-invasive imaging of inflammation and edema formation after kidney transplantation in mice. PLoS ONE. 2016. https://doi.org/10.1371/journal.pone.0162705.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Kimball AB, Summers RM, Turner M, et al. Magnetic resonance imaging detection of occult skin and subcutaneous abnormalities in juvenile dermatomyositis: Implications for diagnosis and therapy. Arthritis Rheum. 2000;43(8):1866–73. https://doi.org/10.1002/1529-0131(200008)43:8%3c1866::AID-ANR24%3e3.0.CO;2-6.

    Article  CAS  PubMed  Google Scholar 

  162. Kociemba A, Pyda M, Katulska K, et al. Comparison of diffusion-weighted with T2-weighted imaging for detection of edema in acute myocardial infarction. J Cardiovasc Magn Reson. 2013;15(1):90. https://doi.org/10.1186/1532-429X-15-90.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Okayama S, Uemura S, Saito Y. Detection of infarct-related myocardial edema using cardiac diffusion-weighted magnetic resonance imaging. Int J Cardiol. 2009;133(1):e20–1. https://doi.org/10.1016/j.ijcard.2007.08.096.

    Article  PubMed  Google Scholar 

  164. Bettoni J, Balédent O, Petruzzo P, et al. Role of flow magnetic resonance imaging in the monitoring of facial allotransplantations: preliminary results on graft vasculopathy. Int J Oral Maxillofac Surg. 2020;49(2):169–75. https://doi.org/10.1016/j.ijom.2019.05.003.

    Article  CAS  PubMed  Google Scholar 

  165. Neugarten J, Golestaneh L. Blood oxygenation level-dependent MRI for assessment of renal oxygenation. Int J Nephrol Renov Dis. 2014;7:421–35. https://doi.org/10.2147/IJNRD.S42924.

    Article  Google Scholar 

  166. Stankovic Z, Allen BD, Garcia J, Jarvis KB, Markl M. 4D flow imaging with MRI. Cardiovasc Diagn Ther. 2014;4(2):173–92. https://doi.org/10.3978/j.issn.2223-3652.2014.01.02.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Alvarez A, Martinez V, Pizarro G, Recio M, Cabrera JÁ. Clinical use of 4D flow MRI for quantification of aortic regurgitation. Open Heart. 2020;7(1): e001158. https://doi.org/10.1136/openhrt-2019-001158.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Turpin J, Unadkat P, Thomas J, et al. Portable magnetic resonance imaging for ICU patients. Crit Care Explor. 2020. https://doi.org/10.1097/CCE.0000000000000306.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Kueckelhaus M, Turk M, Kumamaru KK, et al. Transformation of face transplants: volumetric and morphologic graft changes resemble aging after facial allotransplantation. Am J Transplant. 2016;16(3):968–78. https://doi.org/10.1111/ajt.13544.

    Article  CAS  PubMed  Google Scholar 

  170. Kaufman CL, Kanitakis J, Weissenbacher A, et al. Defining chronic rejection in vascularized composite allotransplantation. SAGE Open Med. 2020. https://doi.org/10.1177/2050312120940421.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Mundinger GS, Drachenberg CB. Chronic rejection in vascularized composite allografts. Curr Opin Organ Transplant. 2014;19(3):309–14. https://doi.org/10.1097/MOT.0000000000000073.

    Article  CAS  PubMed  Google Scholar 

  172. Ballestín A, Casado JG, Abellán E, et al. Ischemia-reperfusion injury in a rat microvascular skin free flap model: a histological, genetic, and blood flow study. PLoS ONE. 2018;13(12): e0209624. https://doi.org/10.1371/journal.pone.0209624.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Iuchi T, Kobayashi M, Tsuchiya S, et al. Objective assessment of leg edema using ultrasonography with a gel pad. PLoS ONE. 2017. https://doi.org/10.1371/journal.pone.0182042.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Kogo H, Murata J, Murata S, Higashi T. Validity of a new quantitative evaluation method that uses the depth of the surface imprint as an indicator for pitting edema. PLoS ONE. 2017;12(1): e0170810. https://doi.org/10.1371/journal.pone.0170810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Pitre J, Vollmer A, Ophir J, Ammann JJ, Weitzel WF, Bull JL. Design and testing of a single-element ultrasound viscoelastography system for point-of-care edema quantification. Ultrasound Med Biol. 2016;42(9):2209–19. https://doi.org/10.1016/j.ultrasmedbio.2016.04.013.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Bragos R, Gámez X, Cairó J, Riu PJ, Gòdia F. Biomass monitoring using impedance spectroscopy. Ann N Y Acad Sci. 1999;873(1):299–305. https://doi.org/10.1111/j.1749-6632.1999.tb09477.x.

    Article  CAS  PubMed  Google Scholar 

  177. Canali C, Heiskanen A, Muhammad HB, et al. Bioimpedance monitoring of 3D cell culturing–complementary electrode configurations for enhanced spatial sensitivity. Biosens Bioelectron. 2015;63:72–9. https://doi.org/10.1016/j.bios.2014.07.020.

    Article  CAS  PubMed  Google Scholar 

  178. González-Araiza JR, Ortiz-Sánchez MC, Vargas-Luna FM, Cabrera-Sixto JM. Application of electrical bio-impedance for the evaluation of strawberry ripeness. Int J Food Prop. 2017;20(5):1044–50. https://doi.org/10.1080/10942912.2016.1199033.

    Article  Google Scholar 

  179. Amini M, Hisdal J, Kalvøy H. Applications of bioimpedance measurement techniques in tissue engineering. J Electr Bioimpedance. 2018;9(1):142–58. https://doi.org/10.2478/joeb-2018-0019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Dylke ES, Ward LC. Three decades of bioelectrical impedance spectroscopy in lymphedema assessment: an historical perspective. Lymphat Res Biol. 2021;19(3):206–14. https://doi.org/10.1089/lrb.2020.0085.

    Article  PubMed  Google Scholar 

  181. Liang ZY, Long X, Yu NZ, Huang JZ. Diagnostic workup of lymphedema. Plast Aesthetic Res. 2019. https://doi.org/10.20517/2347-9264.2019.33.

    Article  Google Scholar 

  182. Payne SC, Alexandrovics J, Thomas R, Shepherd RK, Furness JB, Fallon JB. Transmural impedance detects graded changes of inflammation in experimental colitis. R Soc Open Sci. 2020. https://doi.org/10.1098/rsos.191819.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Yang B, Li B, Xu C, et al. Comparison of electrical impedance tomography and intracranial pressure during dehydration treatment of cerebral edema. NeuroImage Clin. 2019;23: 101909. https://doi.org/10.1016/j.nicl.2019.101909.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Huang X, Cheng H, Chen K, et al. Epidermal impedance sensing sheets for precision hydration assessment and spatial mapping. IEEE Trans Biomed Eng. 2013;60(10):2848–57. https://doi.org/10.1109/TBME.2013.2264879.

    Article  PubMed  Google Scholar 

  185. Dastjerdi HM, Soltanzadeh R, Rabbani H. Designing and implementing bioimpedance spectroscopy device by measuring impedance in a mouse tissue. J Med Signals Sens. 2013;3(3):187–94.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Kenworthy P, Grisbrook TL, Phillips M, et al. Bioimpedance spectroscopy: a technique to monitor interventions for swelling in minor burns. Burns. 2017;43(8):1725–35. https://doi.org/10.1016/j.burns.2017.04.022.

    Article  PubMed  Google Scholar 

  187. Haemmerich D, Staelin ST, Tsai JZ, Tungjitkusolmun S, Mahvi DM, Webster JG. In vivoelectrical conductivity of hepatic tumours. Physiol Meas. 2003;24(2):251–60. https://doi.org/10.1088/0967-3334/24/2/302.

    Article  PubMed  Google Scholar 

  188. Halonen S, Kari J, Ahonen P, Kronström K, Hyttinen J. Real-time bioimpedance-based biopsy needle can identify tissue type with high spatial accuracy. Ann Biomed Eng. 2019;47(3):836–51. https://doi.org/10.1007/s10439-018-02187-9.

    Article  PubMed  Google Scholar 

  189. Halter RJ, Schned A, Heaney J, Hartov A, Schutz S, Paulsen KD. Electrical impedance spectroscopy of benign and malignant prostatic tissues. J Urol. 2008;179(4):1580–6. https://doi.org/10.1016/j.juro.2007.11.043.

    Article  PubMed  Google Scholar 

  190. Halter RJ, Hartov A, Heaney JA, Paulsen KD, Schned AR. Electrical impedance spectroscopy of the human prostate. IEEE Trans Biomed Eng. 2007;54(7):1321–7. https://doi.org/10.1109/TBME.2007.897331.

    Article  PubMed  Google Scholar 

  191. Mahara A, Khan S, Murphy EK, Schned AR, Hyams ES, Halter RJ. 3D microendoscopic electrical impedance tomography for margin assessment during robot-assisted laparoscopic prostatectomy. IEEE Trans Med Imaging. 2015;34(7):1590–601. https://doi.org/10.1109/TMI.2015.2407833.

    Article  PubMed  Google Scholar 

  192. Kalvøy H, Frich L, Grimnes S, Martinsen ØG, Hol PK, Stubhaug A. Impedance-based tissue discrimination for needle guidance. Physiol Meas. 2009;30(2):129–40. https://doi.org/10.1088/0967-3334/30/2/002.

    Article  PubMed  Google Scholar 

  193. McRae DA, Esrick MA, Mueller SC. Changes in the noninvasive, in vivo electrical impedance of three xenografts during the necrotic cell-response sequence. Int J Radiat Oncol. 1999;43(4):849–57. https://doi.org/10.1016/S0360-3016(98)00487-8.

    Article  CAS  Google Scholar 

  194. Shiffman CA. Adverse effects of near current-electrode placement in non-invasive bio-impedance measurements. Physiol Meas. 2013;34(11):1513–29. https://doi.org/10.1088/0967-3334/34/11/1513.

    Article  CAS  PubMed  Google Scholar 

  195. York SL, Ward LC, Czerniec S, Lee MJ, Refshauge KM, Kilbreath SL. Single frequency versus bioimpedance spectroscopy for the assessment of lymphedema. Breast Cancer Res Treat. 2008;117(1):177. https://doi.org/10.1007/s10549-008-0090-6.

    Article  PubMed  Google Scholar 

  196. Seoane F, Macías R, Bragós R, Lindecrantz K. Simple voltage-controlled current source for wideband electrical bioimpedance spectroscopy: circuit dependences and limitations. Meas Sci Technol. 2011;22(11): 115801. https://doi.org/10.1088/0957-0233/22/11/115801.

    Article  CAS  Google Scholar 

  197. Dean DA, Ramanathan T, Machado D, Sundararajan R. Electrical impedance spectroscopy study of biological tissues. J Electrost. 2008;66(3):165–77. https://doi.org/10.1016/j.elstat.2007.11.005.

    Article  CAS  Google Scholar 

  198. Epstein BR, Foster KR. Anisotropy in the dielectric properties of skeletal muscle. Med Biol Eng Comput. 1983;21(1):51. https://doi.org/10.1007/BF02446406.

    Article  CAS  PubMed  Google Scholar 

  199. Naranjo-Hernández D, Reina-Tosina J, Min M. Fundamentals, recent advances, and future challenges in bioimpedance devices for healthcare applications. J Sens N Y. 2019. https://doi.org/10.1155/2019/9210258.

    Article  Google Scholar 

  200. Schneider M, Cardones ARG, Selim MA, Cendales LC. Vascularized composite allotransplantation: a closer look at the banff working classification. Transpl Int. 2016;29(6):663–71. https://doi.org/10.1111/tri.12750.

    Article  PubMed  Google Scholar 

  201. Brouhard BH, Carvajal HF, Linares HA. Burn edema and protein leakage in the rat: I. Relationship to time of injury. Microvasc Res. 1978;15(2):221–8. https://doi.org/10.1016/0026-2862(78)90020-1.

    Article  CAS  PubMed  Google Scholar 

  202. Frueh FS, Körbel C, Gassert L, et al. High-resolution 3D volumetry versus conventional measuring techniques for the assessment of experimental lymphedema in the mouse hindlimb. Sci Rep. 2016. https://doi.org/10.1038/srep34673.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Edwick DO, Hince DA, Rawlins JM, Wood FM, Edgar DW. Bioimpedance spectroscopy is a valid and reliable measure of edema following hand burn injury (part 1—method validation). J Burn Care Res. 2020;41(4):780–7. https://doi.org/10.1093/jbcr/iraa071.

    Article  PubMed  Google Scholar 

  204. Dirks ML, Wall BT, Snijders T, Ottenbros CLP, Verdijk LB, van Loon LJC. Neuromuscular electrical stimulation prevents muscle disuse atrophy during leg immobilization in humans. Acta Physiol Oxf Engl. 2014;210(3):628–41. https://doi.org/10.1111/apha.12200.

    Article  CAS  Google Scholar 

  205. Hardy EJ, Hatt J, Doleman B, et al. Post-operative electrical muscle stimulation attenuates loss of muscle mass and function following major abdominal surgery in older adults: a split body randomised control trial. Age Ageing. 2022;51(10):afac234. https://doi.org/10.1093/ageing/afac234.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Geng K, Wang J, Liu P, et al. Electrical stimulation facilitates the angiogenesis of human umbilical vein endothelial cells through MAPK/ERK signaling pathway by stimulating FGF2 secretion. Am J Physiol Cell Physiol. 2019;317(2):C277–86. https://doi.org/10.1152/ajpcell.00474.2018.

    Article  CAS  PubMed  Google Scholar 

  207. ElAbd R, Alabdulkarim A, AlSabah S, Hazan J, Alhalabi B, Thibaudeau S. Role of electrical stimulation in peripheral nerve regeneration: a systematic review. Plast Reconstr Surg Glob Open. 2022;10(3): e4115. https://doi.org/10.1097/GOX.0000000000004115.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Zealear DL, Rodriguez RJ, Kenny T, et al. Electrical stimulation of a denervated muscle promotes selective reinnervation by native over foreign motoneurons. J Neurophysiol. 2002;87(4):2195–9. https://doi.org/10.1152/jn.00451.2001.

    Article  PubMed  Google Scholar 

  209. Al-Majed AA, Neumann CM, Brushart TM, Gordon T. Brief electrical stimulation promotes the speed and accuracy of motor axonal regeneration. J Neurosci. 2000;20(7):2602–8. https://doi.org/10.1523/JNEUROSCI.20-07-02602.2000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Swisher SL, Lin MC, Liao A, et al. Impedance sensing device enables early detection of pressure ulcers in vivo. Nat Commun. 2015;6(1):6575. https://doi.org/10.1038/ncomms7575.

    Article  CAS  PubMed  Google Scholar 

  211. Carlson K, Barbas A, Goldaracena N, Fernandez L, Al-Adra DP. Immunological organ modification during ex vivo machine perfusion: the future of organ acceptance. Transplant Rev. 2020. https://doi.org/10.1016/j.trre.2020.100586.

    Article  Google Scholar 

  212. Lei B, Sleiman M, Cheng Q, et al. Perfusion of vascularized composite allografts with a targeted complement inhibitor protects against brain death induced and ischemia reperfusion injury. Transplantation. 2020;104(S3):S634. https://doi.org/10.1097/01.tp.0000702020.86870.67.

    Article  CAS  Google Scholar 

  213. Mastroianni M, Ng ZY, Goyal R, et al. Topical delivery of immunosuppression to prolong xenogeneic and allogeneic split-thickness skin graft survival. J Burn Care Res. 2018;39(3):363–73. https://doi.org/10.1097/BCR.0000000000000597.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Guo Y, Messner F, Etra JW, et al. Efficacy of single-agent immunosuppressive regimens in a murine model of vascularized composite allotransplantation. Transpl Int. 2020;33(8):948–57. https://doi.org/10.1111/tri.13618.

    Article  CAS  PubMed  Google Scholar 

  215. Sarhane KA, Ibrahim Z, Leto Barone AA, Cooney DS, Lee WPA, Brandacher G. Minimization of immunosuppression and tolerance induction in reconstructive transplantation. Curr Surg Rep. 2013;1(1):40–6. https://doi.org/10.1007/s40137-012-0008-4.

    Article  Google Scholar 

  216. Chang J, Graves SS, Butts-Miwongtum T, Sale GE, Storb R, Mathes DW. Long-term tolerance towards haploidentical vascularized composite allograft transplantation in a canine model using bone marrow or mobilized stem cells. Transplantation. 2016;100(12):e120–7. https://doi.org/10.1097/TP.0000000000001496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Howsare M, Jones CM, Ramirez AM. Immunosuppression maintenance in vascularized composite allotransplantation: what is just right? Curr Opin Organ Transplant. 2017;22(5):463–9. https://doi.org/10.1097/MOT.0000000000000456.

    Article  PubMed  Google Scholar 

  218. Kanitakis J, Jullien D, Petruzzo P, et al. Clinicopathologic features of graft rejection of the first human hand allograft. Transplantation. 2003;76(4):688–93. https://doi.org/10.1097/01.TP.0000079458.81970.9A.

    Article  PubMed  Google Scholar 

  219. Pei G, Xiang D, Gu L, et al. A report of 15 hand allotransplantations in 12 patients and their outcomes in China. Transplantation. 2012;94(10):1052–9. https://doi.org/10.1097/TP.0b013e31826c3915.

    Article  PubMed  Google Scholar 

  220. Siemionow M. The decade of face transplant outcomes. J Mater Sci Mater Med. 2017;28(5):64. https://doi.org/10.1007/s10856-017-5873-z.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the support of Texas Engineering Experiment Station (TEES) through a professorship to AG-E (TEES-246413). This work was supported by the Office of the Assistant Secretary of Defense for Health Affairs, through the Reconstructive Transplant Research Program Technology/Investigator Initiated Research Award under Award No. W81XWH-16-RTRP-IIRA. Opinions, interpretations, conclusions, and recommendations are those of the author and are not necessarily endorsed by the Department of Defense (W81XWH-17-1-0630).

Funding

Texas A and M Engineering Experiment Station, Texas A and M University, TEES-246413, Anthony Guiseppi-Elie, Medical Research and Materiel Command, W81XWH-16-RTRP-IIRA, Warren L. Grayson, W81XWH-17-1-0630, Warren L. Grayson.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Gerald Brandacher, Anthony Guiseppi-Elie or Warren L. Grayson.

Ethics declarations

Competing interests

Prof. Guiseppi-Elie is founder, president, and scientific director of ABTECH Scientific, Inc., manufacturer of microfabricated electrodes and devices used in biomedical diagnostics and the measurement of physiological data. The funding sponsors had no role in the design of the study; in the collection, analysis, or interpretation of data; in the writing of the manuscript, and in the decision to publish the findings.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ton, C., Salehi, S., Abasi, S. et al. Methods of ex vivo analysis of tissue status in vascularized composite allografts. J Transl Med 21, 609 (2023). https://doi.org/10.1186/s12967-023-04379-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-04379-x

Keywords