Next Article in Journal
LARIAT or AtriClip: Complications Profile and Comparison in Patients with Atrial Fibrillations Based on Manufacturer and User Facility Device Experience Database
Previous Article in Journal
Eco-Anxiety: An Evolutionary Line from Psychology to Psychopathology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Biomarkers to Predict Multiorgan Distress Syndrome and Acute Kidney Injury in Critically Ill Surgical Patients

1
Division of Acute Care Surgery, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
2
Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
3
Division of Colorectal Surgery, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
4
Center of Evidence Based Medicine, Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea
5
Division of Esophago-Gastrointestinal Surgery, Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
6
Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
7
Central Research Laboratory, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
8
Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Medicina 2023, 59(12), 2054; https://doi.org/10.3390/medicina59122054
Submission received: 16 September 2023 / Revised: 17 November 2023 / Accepted: 20 November 2023 / Published: 21 November 2023
(This article belongs to the Section Surgery)

Abstract

:
Background and Objectives: Critically ill surgical patients are susceptible to various postoperative complications, including acute kidney injury (AKI) and multiorgan distress syndrome (MODS). These complications intensify patient suffering and significantly increase morbidity and mortality rates. This study aimed to identify the biomarkers for predicting AKI and MODS in critically ill surgical patients. Materials and Methods: We prospectively enrolled critically ill surgical patients admitted to the intensive care unit via the emergency department between July 2022 and July 2023. A total of 83 patients were recruited, and their data were used to analyze MODS. Three patients who showed decreased creatinine clearance at the initial presentation were excluded from the analysis for AKI. Patient characteristics and laboratory parameters including white blood cell (WBC) count, neutrophil count, delta neutrophil index, urine and serum β2-microglobulin, and urine serum mitochondrial DNA copy number (mtDNAcn) were analyzed to determine the reliable biomarker to predict AKI and MODS. Results: The following parameters were independently correlated with MODS: systolic blood pressure (SBP), initial neutrophil count, and platelet count, according to a logistic regression model. The optimal cut-off values for SBP, initial neutrophil count, and platelet count were 113 mmHg (sensitivity 66.7%; specificity 73.9%), 8.65 (X3) (109/L) (sensitivity 72.2%; specificity 64.6%), and 195.0 (X3) (109/L) (sensitivity 66.7%; specificity 81.5%), respectively. According to the logistic regression model, diastolic blood pressure (DBP) and initial urine mtDNAcn were independently correlated with AKI. The optimal cut-off value for DBP and initial urine mtDNAcn were 68.5 mmHg (sensitivity 61.1%; specificity 79.5%) and 1225.6 copies/μL (sensitivity 55.6%; specificity 95.5%), respectively. Conclusions: SBP, initial neutrophil count, and platelet count were independent predictors of MODS in critically ill patients undergoing surgery. DBP and initial urine mtDNAcn levels were independent predictors of AKI in critically ill surgical patients. Large-scale multicenter prospective studies are needed to confirm our results.

1. Introduction

Critically ill surgical patients are susceptible to various postoperative complications, including acute kidney injury (AKI) and multiorgan distress syndrome (MODS). These complications not only intensify patient suffering, but also significantly increase morbidity and mortality rates, compelling the medical community to explore innovative approaches for their early identification and management [1,2]. Biomarkers have emerged as promising candidates for addressing this clinical need.
Early detection of AKI and MODS is pivotal for informed clinical decision making and optimized patient care. Recently, the biological relevance of mitochondrial DNA (mtDNA) in predicting adverse outcomes has been increasingly recognized, particularly in patients in the intensive care unit (ICU) [3,4,5,6]. Mitochondrial integrity plays a pivotal role in AKI pathophysiology. In various forms of AKI, early pathological alterations are evident within the renal tubular epithelium, including reduced mitochondrial abundance, organelle swelling, and fragmentation [7]. The impaired state of these mitochondria causes kidney damage by producing detrimental reactive oxygen species and releasing mtDNA. The release of mtDNA can activate innate immune responses via the Toll-like receptor-9 (TLR9) pathway [8]. Moreover, mtDNA acts as a damage-associated molecular pattern initiator [9] that can drive molecular processes leading to inflammatory responses and organ injuries [9,10,11].
β2-microglobulin (β2-MG) has also been investigated recently as a predictive marker, in particular for AKI [12,13]. β2-MG is released into circulation at a constant rate, freely filtered by the glomeruli, and completely reabsorbed and catabolized in the renal tubules. Serum β2-MG levels are independent of muscle mass and start increasing early during kidney failure. Because of these properties, serum β2-MG has been proposed as a candidate marker to assess kidney function. Tubular injury leads to decreased reabsorption of β2-MG and tubular enzymes, leading to elevated urinary concentrations. In addition, a few studies reported that β2-MG was associated with severe inflammation [14,15].
Based on these findings, we hypothesized that urine mtDNA, circulating cell-free mtDNA levels, serum β2-MG, and urinary β2-MG would be associated with AKI and MODS and would improve risk prediction in critically ill surgical patients. Although many studies have investigated the effectiveness of mtDNA as a predictive marker, studies conducted on patients admitted to the intensive care unit because of acute surgical illnesses are rare.
Therefore, this study aimed to test whether circulating cell-free mtDNA, urinary mtDNA copy number (mtDNAcn), serum β2-MG, and urinary β2-MG are useful as biomarkers in critically ill surgical patients. We also investigated other well-known biomarkers, including the delta neutrophil index (DNI), white blood cell (WBC) count, and neutrophil count because of the scarcity of studies on critically ill surgical patients.

2. Materials and Methods

The protocol of this study was registered in clinicaltrials.gov (NCT05458063).

2.1. Patient Selection

This was a prospective, observational study. This study was approved by the Ethics Committee of Wonju Severance Christian hospital (Institutional Review Board No. CR322053). We prospectively enrolled critically ill surgical patients admitted to the ICU via emergency department between July 2022 and July 2023. The exclusion criteria were as follows: (1) age < 18 years; (2) pregnancy; (3) death at initial presentation; (4) history of underlying chronic renal disease; and (5) patients with a previous history of AKI. A total of 120 patients were screened initially. Among them, 83 patients were recruited and their data were used to analyze MODS. Three patients with decreased creatinine clearance were excluded from the AKI risk factor analysis.

2.2. Data Collection and Definition

After obtaining written informed consent, background clinical information and the presence of other comorbid conditions were recorded. The following parameters were recorded prospectively: age; sex; vital signs at the initial presentation including systolic blood pressure (SBP), diastolic blood pressure (DBP), pulse rate (PR), and body temperature (BT); and laboratory parameters at the initial presentation including DNI, WBC, neutrophil, and platelet counts, and levels of creatinine, serum β2-MG, urine β2-MG, hemoglobin, the international normalized ratio (INR), c-reactive protein (CRP), lactate, serum mtDNAcn, and urine mtDNAcn. In addition, DNI, WBC count, neutrophil count, serum and urine β2-MG levels, and serum and urine mtDNAcn on first, second, and third days after admission were recorded to observe daily changes. The sequential organ failure assessment (SOFA) score was calculated and collected prospectively during the ICU stay. After the patients were discharged, outcomes, including AKI, MODS, and mortality, were recorded. MODS was defined as a SOFA score of 6 or higher on two or more consecutive days, at least 48 h after emergency department admission [16]. AKI was defined according to the Acute Kidney Injury Network (AKIN) criteria [17].

2.3. Blood and Urine Sampling, Preparation, and Storage

Blood samples were drawn and transferred into ethylenediamine tetraacetic acid (EDTA)-coated blood collection tubes at the initial presentation. Blood samples were collected at 24, 48, and 72 h after the first collection. The blood samples were sent to the central laboratory within 2 h after every venipuncture, and the EDTA tubes were centrifuged with 1300× g for 10 min at −4 °C. After first centrifugation, 1500 μL supernatant was collected and transferred into a 1.5 mL microtube. The microtube was centrifuged with 4000× g for 10 min at −4 °C, and 1000 μL supernatant was transferred into a new sterile 1.5 mL microtube, and stored at −80 °C in the freezer. Urine samples were drawn and transferred into urine tubes at the initial presentation and at 24, 48, and 72 h after the first collection. The urine samples were also sent to the central laboratory within 2 h after collection, and 1500 μL urine was collected, and transferred into a 1.5 mL microtube. The microtube was centrifuged with 4000× g for 10 min at −4 °C, and 1000 μL supernatant was transferred into a 1.5 mL microtube, and stored at −80 °C in the freezer until analysis. Freeze–thaw cycles were avoided to reduce the phenomenon of DNA fragmentation, and the extracts were not retained for >3 months at −80 °C.

2.4. DNA Isolation from Plasma and Urine

DNA extraction from plasma and urine was performed using a DNA mini kit (#51306; Qiagen, Hilden, Germany) following the manufacturer’s protocol. We incubated samples with lysis buffer (included in the kit) and proteinase K at 56 °C for 10 min. After adding ethanol (96–100%), the mixture was applied to a mini spin column (included in the kit) and centrifuged, and the filtrate was removed. The pellet in the mini-spin column was washed twice with two washing buffers (included in the kit). DNA was eluted in 50 μL of distilled water.

2.5. Primers and Quantitative Polymerase Chain Reaction (qPCR)

DNA was extracted from 200 µL (sample volume) plasma and urine using the QIAamp DNA mini kit (#51306; Qiagen, Germany). Purified DNA was eluted in 50 µL (elution volume) distilled water. The mtDNAcn was measured by a SYBR Green dye-based qPCR assay using Quantstudio 6 Flex real time PCR (Applied Biosystems, Waltham, MA, USA). The primer sequences were as follows: H-Mito Forward: 5′-CACTTTCCACACAGACATCA-3′, Reverse: 5′-TGGTTAGGCTGGTGTTAGGG-3′ [18]. A dilution series from 1 × 108 copies/μL to 1 × 102 copies/μL consisting of the purified PCR product from a healthy control participant, not participating in the present study, was constructed and used to create a standard curve. Concentrations were converted to copy number using the formula: mol/g 6 molecules/mol = molecules/g via a DNA copy number calculator (http://cels.uri.edu/gsc/cndna.html (accessed on 16 May 2023); University of Rhode Island Genomics and Sequencing Center).
The thermal profile for detecting mtDNA using Quantinova SYBR Green PCR kit (Qiagen, Germany) was analyzed as follows: an initiation step for 2 min at 60 °C was followed by an initial denaturation step for 10 min at 95 °C and a further step consisting of 40 cycles for 20 s at 95 °C and for 10 s at 60 °C. The representative standard curves, dissociation curves, and amplification plots are shown in Figure 1.
All samples were analyzed in triplicate, and a no-template control was included in each analysis. The mtDNA levels in all plasma analyses were expressed in copies per microliter of plasma based on the following calculations [19]:
C o p y   N u m b e r   p e r   μ L = C o p y   n u m b e r   p e r   2   μ L   r e a c t i o n × E l u t i o n   v o l u m e   μ L   q P C R   r e a c t i o n   v o l u m e   μ L × 1 S a m p l e   v o l u m e   μ L

2.6. Statistical Analysis

Statistical analyses were performed using R statistical software (version 4.1.0; R Foundation for Statistical Computing, Vienna, Austria). Continuous variables are presented as the mean and standard deviation or median and range, and a comparative analysis was conducted using the independent-samples t-test. The Mann–Whitney U-test was performed for continuous variables that were non-normally distributed. Chi-squared and Fisher’s exact tests were used for comparative analysis of categorical variables. Multivariate analysis was performed using logistic regression to identify independent risk factors. A receiver-operating characteristic (ROC) curve was constructed and the Youden index method was used to determine the optimal cut-off values for predicting mortality. Statistical significance was set at p-value < 0.05.

3. Results

3.1. Baseline Characteristics in the Enrolled Patients

Mean age in all patients was 63.3 ± 17.3 years, and the male:female ratio was 56:27. A total of 30 patients (36.1%) had hypertension and 18 patients (21.7%) had diabetes mellitus. The most common diagnosis was pan-peritonitis (41 patients [49.4%]), followed by hemoperitoneum (16 patients [19.2%]). Surgical treatment was performed in 76 patients (91.6%) (Table 1).

3.2. Patient Characteristics between Patients without and with MODS

Mean age (62.0 ± 18.4 vs. 68.0 ± 11.7 years, p = 0.317) and the ratio of male sex (42 [64.6%] vs. 14 [77.8%], p = 0.441) were not significantly different between both groups. SBP (126.1 ± 26.5 vs. 98.2 ± 31.5 mmHg, p < 0.001) and DBP (70.6 ± 16.4 vs. 55.2 ± 15.4 mmHg, p = 0.001) were significantly lower in patients with MODS. Initial DNI (4.1 ± 8.4% vs. 9.8 ± 12.2%, p = 0.016), WBC (12.8 ± 5.6 vs. 10.0 ± 4.9 (X3) (109/L), 0.046), neutrophil (10.8 ± 5.2 vs. 7.8 ± 3.8 (X3) (109/L), p = 0.028), creatinine (0.8 ± 0.3 vs. 1.2 ± 0.3 mg/dL, p < 0.001), serum β2-MG (2.5 ± 2.0 vs. 4.2 ± 4.1 mg/L, p = 0.008), hemoglobin (13.1 ± 2.3 vs. 11.8 ± 1.9 g/dL, p = 0.026), platelet (261.0 ± 107.3 vs. 188.9 ± 81.6 (X3) (109/L), p = 0.004), INR (1.0 ± 0.1 vs. 1.2 ± 0.2, p < 0.001), and lactate (2.5 ± 1.9 vs. 4.7 ± 3.9 mmol/L, p = 0.005) were significantly different between both groups. Initial urine mtDNAcn was significantly higher in patients with MODS (1067.5 ± 1575.8 vs. 3800.4 ± 7298.9 copies/μL, p = 0.032). The highest SOFA score was significantly higher in patients with MODS (2.0 ± 1.6 vs. 8.1 ± 2.0, p < 0.001). Hospital duration of stay (DOS) (15.8 ± 10.1 vs. 28.7 ± 22.0 days, p = 0.008) and ICU DOS (3.5 ± 3.7 vs. 9.1 ± 7.7 days, p = 0.001) were significantly higher in patients with MODS. Mortality was more frequent in patients with MODS (0 [0%] vs. 5 [27.8%], p < 0.001) (Table 2).

3.3. Independent Risk Factors to Predict MODS in Critically Ill Surgical Patients

A logistic regression model was performed to determine independent risk factors, including age, sex, SBP, DBP, initial DNI, WBC, neutrophil, creatinine, serum β2-MG, hemoglobin, platelet, INR, lactate, and initial urine mtDNAcn. The following were independently correlated with MODS: SBP (odds ratio [OR] 0.97, 95% confidence interval [CI] 0.94–1.00, p = 0.025], initial neutrophil count [OR 0.84, 95% CI 0.72–0.98, p = 0.024], and initial platelet count [OR 0.99, 0.98–1.00, p = 0.036] (Table 3).

3.4. Optimal Cut-Off Value of SBP and Initial Neutrophil and Platelet Counts to Predict MODS in Critically Ill Surgical Patients

The ROC curves of SBP and initial neutrophil and platelet counts were calculated to predict MODS in critically ill patients undergoing surgery. The areas under the curve (AUC) of SBP and initial neutrophil and platelet counts were 0.750 (95% CI 0.606–0.877), 0.670 (95% CI 0.539–0.801), and 0.724 (0.570–0.877), respectively. The optimal cut-off values for SBP and initial neutrophil and platelet counts were 113 mmHg (sensitivity 66.7%; specificity 73.9%), 8.65 (X3) (109/L) (sensitivity 72.2%; specificity 64.6%), and 195.0 (X3) (109/L) (sensitivity 66.7%; specificity 81.5%), respectively (Table 4 and Figure 2).

3.5. Patient Characteristics between Patients without and with AKI

Mean age (59.4 ± 17.9 vs. 66.2 ± 15.5 years, p = 0.072) and the ratio of male sex (30 [68.2%] vs. 24 [66.7%], p = 1.000) were not significantly different between both groups. SBP (127.2 ± 19.3 vs. 112.8 ± 37.9 mmHg, p = 0.044) and DBP (74.4 ± 13.8 vs. 58.6 ± 17.8 mmHg, p < 0.001) were significantly lower in patients with AKI. Initial DNI (2.7 ± 4.8% vs. 7.3 ± 11.0%, p = 0.035), WBC (15.1 ± 13.4 vs. 11.6 ± 5.9 (X3) (109/L), p = 0.085), neutrophil (11.2 ± 4.7 vs. 9.3 ± 5.2 (X3) (109/L), p = 0.045), creatinine (0.8 ± 0.2 vs. 1.0 ± 0.4 mg/dL, p = 0.006), serum β2-MG (2.1 ± 1.4 vs. 3.4 ± 3.4 mg/L, p = 0.020), hemoglobin (13.4 ± 2.1 vs. 12.2 ± 2.3 g/dL, p = 0.011), platelet (263.6 ± 93.6 vs. 222.6 ± 120.6 (X3) (109/L), p = 0.016), INR (1.0 ± 0.1 vs. 1.1 ± 0.2, p = 0.007), and lactate (2.2 ± 1.5 vs. 3.9 ± 3.3 mmol/L, p = 0.004) were significantly different between both groups. Initial urine mtDNAcn was significantly higher in patients with AKI (561.4 ± 349.9 vs. 3001.8 ± 5424.5 copies/μL, p = 0.001). The highest SOFA score was significantly higher in patients with AKI (2.2 ± 2.0 vs. 4.5 ± 3.7, p = 0.004). Hospital DOS (15.9 ± 14.7 vs. 21.6 ± 14.3 days, p = 0.007) and ICU DOS (3.1 ± 3.7 vs. 6.5 ± 6.4 days, p = 0.010) were significantly higher in patients with AKI. MODS (4 [9.1%] vs. 14 [38.9%], p = 0.004) and mortality were more common in patients with AKI (0 [0%] vs. 5 [13.9%], p = 0.037) (Table 5).

3.6. Independent Risk Factors to Predict AKI in Critically Ill Surgical Patients

A logistic regression model was performed to determine independent risk factors, including age, sex, SBP, DBP, initial DNI, neutrophil, creatinine, serum β2-MG, hemoglobin, platelet, INR, lactate, and initial urine mtDNAcn. The following were independently correlated with AKI: DBP (OR 0.90, 95% CI 0.84–0.96, p = 0.001) and initial urine mtDNAcn (OR 1.00, 95% CI 1.00–1.00, p = 0.010) (Table 6).

3.7. Optimal Cut-Off Value of DBP and Initial Urine mtDNAcn to Predict AKI in Critically Ill Surgical Patients

The ROC curves of DBP and initial urine mtDNAcn were calculated to predict AKI in critically ill surgical patients. The AUC of DBP and initial urine mtDNAcn were 0.755 (95% CI 0.647–0.864) and 0.718 (95% CI 0.590–0.845), respectively. The optimal cut-off values for DBP and initial urine mtDNAcn were 68.5 mmHg (sensitivity 61.1%; specificity 79.5%) and 1225.6 (sensitivity, 55.6%; specificity, 95.5%), respectively (Table 7 and Figure 3).

4. Discussion

In this study, we found that SBP and initial neutrophil and platelet counts were independently associated with MODS in critically ill patients undergoing surgery. The optimal cut-off values for SBP and initial neutrophil and platelet counts were 113 mmHg, 8.65 (X3)(109/L), and 195.0 (X3)(109/L), respectively. In addition, DBP and initial urine mtDNAcn levels were independently associated with AKI in critically ill surgical patients. The optimal cut-off values for DBP and initial urine mtDNAcn were 68.5 mmHg and 1225.6 copies/μL, respectively.
Consistent with our findings, several studies have reported an established relationship between SBP and MODS occurrence across various clinical contexts [20,21]. This underscores the importance of maintaining optimal perfusion and effectively managing blood pressure in critically ill patients. Transient episodes of hypotension can precipitate inadequate oxygen delivery to vital organs, initiating a cascade of events that may ultimately culminate in MODS. Early recognition and prompt management of hypotension are imperative for preventing severe medical complications.
Polymorphonuclear leukocytes, monocytes, macrophages, dendritic cells, and natural killer cells are pivotal contributors to the cellular immune response following infection or trauma [22,23]. Neutrophils are attracted to the site of injury under the influence of cytokines, such as interleukin (IL)-8, where they actively participate in the immune response by combatting pathogens and assisting in the removal of damaged tissue. Additionally, these cells play a role in the initiation of molecules such as tumor necrosis factor (TNF)-α, IL-8, platelet-activating factor, and anaphylatoxin (C5a). This sequence of events leads to an increased inflammatory response, which triggers the activation and recruitment of polymorphonuclear leukocytes. This, in turn, contributes to the onset of the systemic inflammatory response syndrome and MODS [22,23,24]. Recruitment of polymorphonuclear cells leads to neutrophilia. This period can be considered a “vulnerability window” during which a subsequent event, often referred to as a “second hit”, can trigger the onset of MODS [24,25]. In patients who develop MODS, initial neutrophilia is followed by neutropenia following trauma or infection [26]. Therefore, patients with neutropenia at initial presentation may progress to MODS, as shown in our study.
Thrombocytopenia at initial presentation is most likely caused by the loss or consumption of platelets or sepsis in critically ill surgical patients. Thrombocytopenia in patients with sepsis occurs through several mechanisms. Platelets are activated during sepsis and adhere to the endothelial lining, leading to their sequestration and subsequent breakdown. Additionally, immune-related factors such as non-specific antibodies associated with platelets and cytokine-driven phagocytosis of platelets can also play a role in the development of thrombocytopenia in sepsis [27]. Therefore, the development of thrombocytopenia indicates progression to more serious adverse events. Similar to our results, previous studies have reported that thrombocytopenia poses an independent risk for adverse events in ICU patients [28,29].
DBP serves as an indicator of the vascular tone. A study has illustrated that elevated levels of naturally occurring inflammatory mediators, such as nitrous oxide and TNF-α, are linked to the progression of severe sepsis, shock, or even mortality [30]. These cytokines and autocrine hormones have vasodilatory properties. Patients with sepsis who exhibit a normal mean SBP and low DBP are in a state of compensated vasodilation, which precedes a more evident cardiovascular collapse. In patients with trauma, there can be an initial mild shock leading to an increase in systemic venous resistance and subsequently higher DBP. However, when severe and ongoing bleeding occurs, resulting in a loss of over 20–30% of the total blood volume, systemic venous resistance may decrease, leading to failure of vascular compensation [31]. This, in turn, causes a decline in DBP. Therefore, a reduction in DBP in patients with trauma results in a substantial loss of blood volume. In other words, a lower DBP reflects a lower effective circulating volume at initial presentation and a high prevalence of AKI.
The mtDNA has a damage-associated molecular pattern (DAMP). In a landmark study in 2004, Collins et al. [32]. found that injecting mtDNA into the joints of mice resulted in localized inflammation. Many studies have investigated the mechanism by which mtDNA affects the inflammatory cascade by acting as a DAMP. First, mtDNA is released into the cytoplasm or extracellular space via various mechanisms in various clinical settings. Various mitochondrial stresses, including bacterial and viral infections or trauma-induced cellular damage, can lead to the release of mtDNA associated with reactive oxygen species. Alternatively, activation of Bcl-2-associated X protein (BAX) and Bcl-2 homologous antagonist/killer (BAK) leads to outer mitochondrial membrane permeabilization (MOMP) and mtDNA release [33]. Pathogen-infected cells often secrete IL-1β due to inflammasome activation. A recent report by Aarreberg et al. [34] discovers a link between IL-1β secretion in infected cells, which can then activate a cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING)-dependent type-I interferon response in surrounding bystander cells. Interestingly, IL-1β stimulation of bystander cells increases mitochondrial mass, decreases mitochondrial membrane potential, and induces mtDNA release.
Once cytoplasmic, mtDNA released from mitochondria can also bind the DNA sensing protein cGAS that catalyzes the production of the secondary messenger 2030 cyclic GMP–AMP (2030cGAMP) from adenosine triphosphate (ATP) and guanosine-5′-triphosphate (GTP). Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) binds to the adaptor molecule STING on the endoplasmic reticulum, leading to the activation of TANK-binding kinase 1 (TBK1). Active TBK1 phosphorylates the transcription factor interferon regulatory factor 3 (IRF3) initiating a type-I interferon response [33]. Elevated levels of type-I interferons can hinder B-cell reactions, possibly inducing the generation of immunosuppressive substances. Additionally, these elevated concentrations diminish the ability of macrophages to respond effectively to interferon-γ activation [35].
Cytoplasmic mtDNA can enter the extracellular environment through necroptosis or platelet aggregation [36]. The released circulating mtDNA is detected by the TLR9 located on the neutrophil surface, which activates p38 mitogen-activated protein kinase (MAPK). Subsequently, diverse transcriptional factors including nuclear factor kappa-light-chain-enhancer of activated B cells is activated via MAPK, and thereby a cellular response is induced. Cellular responses include the expression of cytokines or adhesion molecules that accelerate inflammation and diapedesis of immune effector cells. In addition, released IL-1β also contributes to systemic inflammatory responses.
Therefore, we hypothesized that serum mtDNAcn level may be a predictor of MODS. However, we did not observe any difference in serum mtDNAcn levels between patients with and without MODS. Nakashira et al. [5] reported that circulating mtDNA levels were associated with mortality in medical ICU patients. Harrington et al. [36] reported that 11 out of 16 studies reported a significant association between circulating mtDNA levels and mortality among critically ill patients in their systematic review. These discrepancies between studies on critically ill patients, including that of our study, although our outcome was not mortality, may be due to the indirectness of the participants and small number of participants.
Mitochondria are pivotal contributors to AKI, serving a dual function as the primary energy source for cells and a crucial controller of cell death processes. Initially, alterations in the mitochondrial structure were detected in AKI cases. A primary cause of AKI is ischemia, which leads to a reduction in the number of mitochondria and causes structural changes in these organelles, characterized by swelling and loss of the inner mitochondrial membrane cristae. These changes occur because of diminished ATP levels and a decline in membrane potential [37]. Furthermore, the opening of mitochondrial permeability transition pores (mPTP) is a pivotal event resulting from mitochondrial swelling and dysfunction. This event significantly contributes to AKI progression by releasing proapoptotic substances such as cytochrome c, which can trigger apoptosis in renal cells [38]. Second, an imbalance in the regulation of mitochondrial fission and fusion has been linked to AKI progression. In this context, dynamin-related protein 1, which is responsible for controlling mitochondrial fission, exhibits swift activation, whereas mitofusin and optic atrophy 1, which oversee mitochondrial fusion, experience a reduction in their levels during AKI. This process leads to fragmentation of mitochondria [39]. Third, the released mtDNA has the ability to bind to the DNA-sensing protein cGAS, triggering a sequence of events, as described earlier. Fourth, oxidative stress produces mitochondrial reactive oxygen species, which can destroy renal cells [40]. Finally, the release of mtDNA as one of DAMPs from mitochondria into the extracellular space accelerates a vicious spiral of renal cellular damage [41]. Urinary mtDNA levels can be elevated because of AKI-related mitochondrial dysfunction. Hu et al. [41] reported that urinary mtDNA levels were associated with new-onset AKI in surgical ICU patients, which is consistent with our results. However, Whitaker et al. [42] showed that urinary mtDNA levels did not differ between AKI and non-AKI groups after cardiac surgery. Additional research is needed to gain a more comprehensive understanding of the predictive potential of urinary mtDNA for AKI as variations and inconsistencies persist among studies.
Although serum β2-MG was not an independent predictor of AKI and MODS (p = 0.053) in the multivariate analysis in this study, it could be a candidate of predictors for AKI and MODS. β2-MG is filtered by glomeruli and subsequently reabsorbed in the proximal tubule. Serum β2-MG is independent of muscle mass and starts increasing early in kidney failure. β2-MG is present in all nucleated cells [43] and is not affected by sex or age [44]. These properties may make β2-MG an ideal endogenous biomarker for estimating the glomerular filtration rate. Tubular injury leads to decreased reabsorption of β2-MG and tubular enzymes, leading to elevated urinary concentrations. Thus, these markers may act as functional markers of tubular damage and fibrosis. In addition, β2-MG may be related to inflammation. Although not fully understood, β2-MG may induce IL-1β and IL-18 release by macrophages in a caspase-1- and nod-like receptor family pyrin-domain-containing 3-dependent manner [45]. Further investigation is necessary to determine the association between β2-MG and adverse outcomes.
Our study has several limitations. First, our sample size and the number of patients with MODS were small (n = 18). Second, several methodological factors may have influenced our results. For example, although we prepared samples within 2 h of collection, we cannot rule out the possibility that slight differences in the timing of sample collection could influence the associations within the groups (AKI vs. no AKI, MODS vs. no MODS). Moreover, slight differences in mtDNA measurements can occur throughout the process, from sample collection to qPCR. Third, although we considered many variables for the multivariate analysis, some possible factors could have been missed. Finally, our study included surgical patients admitted to the ICU, both trauma and non-trauma patients. The mechanism of MODS development may differ between patients, depending on whether it is related to infection or sterile inflammation. This may have influenced the results of this study.

5. Conclusions

Despite these limitations, our study is meaningful because it is a rare prospective observational study that comprehensively analyzed biomarkers to predict MODS and AKI in critically ill surgical patients. SBP, initial neutrophil count, and platelet count were independent predictors of MODS in critically ill patients. In addition, we found that DBP and initial urine mtDNAcn levels were independent predictors of AKI in critically ill surgical patients. Large-scale multicenter prospective studies are needed to confirm our results.

Author Contributions

Conceptualization, I.S.S., D.K.K., S.A., S.C.G., M.H.K., M.H.R., C.-S.K., J.H.S., K.K. and H.R.; methodology, I.S.S., D.K.K., M.H.R., C.-S.K., J.H.S., K.K. and H.R.; software, I.S.S., M.H.R., C.-S.K., J.H.S., K.K. and H.R.; validation, I.S.S., D.K.K., S.A., S.C.G., M.H.K., K.K. and H.R.; Formal analysis, I.S.S. and K.K.; investigation, I.S.S., D.K.K., H.R., C.-S.K., J.H.S. and K.K.; resources, I.S.S. and K.K.; data curation, I.S.S. and K.K.; writing—original draft preparation, I.S.S. and K.K.; writing—review and editing, I.S.S., D.K.K., S.A., S.C.G., M.H.K., M.H.R., C.-S.K., J.H.S., K.K. and H.R.; visualization, I.S.S. and K.K.; supervision, C.-S.K., J.H.S., K.K. and H.R.; project administration, I.S.S., D.K.K. and K.K.; funding acquisition, I.S.S. and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Ministry of Education, grant number 2022R1I1A1A0106951112.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of Wonju Severance Christian hospital (Institutional Review Board No. CR322053, 7 July 2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in this study.

Data Availability Statement

Data can be made available upon request.

Acknowledgments

We would like to thank Sang Mi Lee for her efforts and dedication to this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rewa, O.; Bagshaw, S.M. Acute kidney injury-epidemiology, outcomes and economics. Nat. Rev. Nephrol. 2014, 10, 193–207. [Google Scholar] [CrossRef]
  2. Umegaki, T.; Ikai, H.; Imanaka, Y. The impact of acute organ dysfunction on patients’ mortality with severe sepsis. J. Anaesthesiol. Clin. Pharmacol. 2011, 27, 180–184. [Google Scholar] [CrossRef]
  3. Pyle, A.; Burn, D.J.; Gordon, C.; Swan, C.; Chinnery, P.F.; Baudouin, S.V. Fall in circulating mononuclear cell mitochondrial DNA content in human sepsis. Intensive Care Med. 2010, 36, 956–962. [Google Scholar] [CrossRef]
  4. Che, R.; Yuan, Y.; Huang, S.; Zhang, A. Mitochondrial dysfunction in the pathophysiology of renal diseases. Am. J. Physiol. Renal Physiol. 2014, 306, F367–F378. [Google Scholar] [CrossRef]
  5. Nakahira, K.; Kyung, S.Y.; Rogers, A.J.; Gazourian, L.; Youn, S.; Massaro, A.F.; Quintana, C.; Osorio, J.C.; Wang, Z.; Zhao, Y.; et al. Circulating mitochondrial DNA in patients in the icu as a marker of mortality: Derivation and validation. PLoS Med. 2013, 10, e1001577, discussion e1001577. [Google Scholar] [CrossRef]
  6. Ho, P.W.; Pang, W.F.; Luk, C.C.; Ng, J.K.; Chow, K.M.; Kwan, B.C.; Li, P.K.; Szeto, C.C. Urinary mitochondrial DNA level as a biomarker of acute kidney injury severity. Kidney Dis. 2017, 3, 78–83. [Google Scholar] [CrossRef]
  7. Tran, M.; Tam, D.; Bardia, A.; Bhasin, M.; Rowe, G.C.; Kher, A.; Zsengeller, Z.K.; Akhavan-Sharif, M.R.; Khankin, E.V.; Saintgeniez, M.; et al. Pgc-1alpha promotes recovery after acute kidney injury during systemic inflammation in mice. J. Clin. Investig. 2011, 121, 4003–4014. [Google Scholar] [CrossRef]
  8. Emma, F.; Montini, G.; Parikh, S.M.; Salviati, L. Mitochondrial dysfunction in inherited renal disease and acute kidney injury. Nat. Rev. Nephrol. 2016, 12, 267–280. [Google Scholar] [CrossRef]
  9. Krysko, D.V.; Agostinis, P.; Krysko, O.; Garg, A.D.; Bachert, C.; Lambrecht, B.N.; Vandenabeele, P. Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol. 2011, 32, 157–164. [Google Scholar] [CrossRef]
  10. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial damps cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef]
  11. Sursal, T.; Stearns-Kurosawa, D.J.; Itagaki, K.; Oh, S.Y.; Sun, S.; Kurosawa, S.; Hauser, C.J. Plasma bacterial and mitochondrial DNA distinguish bacterial sepsis from sterile systemic inflammatory response syndrome and quantify inflammatory tissue injury in nonhuman primates. Shock 2013, 39, 55–62. [Google Scholar] [CrossRef]
  12. Cheung, A.K.; Rocco, M.V.; Yan, G.; Leypoldt, J.K.; Levin, N.W.; Greene, T.; Agodoa, L.; Bailey, J.; Beck, G.J.; Clark, W.; et al. Serum beta-2 microglobulin levels predict mortality in dialysis patients: Results of the hemo study. J. Am. Soc. Nephrol. 2006, 17, 546–555. [Google Scholar] [CrossRef]
  13. Wang, R.; Hu, H.; Hu, S.; He, H.; Shui, H. β2-microglobulin is an independent indicator of acute kidney injury and outcomes in patients with intracerebral hemorrhage. Medicine 2020, 99, e19212. [Google Scholar] [CrossRef]
  14. Yilmaz, B.; Koklu, S.; Yuksel, O.; Arslan, S. Serum beta 2-microglobulin as a biomarker in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 10916–10920. [Google Scholar] [CrossRef]
  15. Yang, J.; Qian, J.; Wezeman, M.; Wang, S.; Lin, P.; Wang, M.; Yaccoby, S.; Kwak, L.W.; Barlogie, B.; Yi, Q. Targeting beta2-microglobulin for induction of tumor apoptosis in human hematological malignancies. Cancer Cell 2006, 10, 295–307. [Google Scholar] [CrossRef]
  16. Kong, T.; Park, Y.S.; Lee, H.S.; Kim, S.; Lee, J.W.; You, J.S.; Chung, H.S.; Park, I.; Chung, S.P. The delta neutrophil index predicts development of multiple organ dysfunction syndrome and 30-day mortality in trauma patients admitted to an intensive care unit: A retrospective analysis. Sci. Rep. 2018, 8, 17515. [Google Scholar] [CrossRef]
  17. Mehta, R.L.; Kellum, J.A.; Shah, S.V.; Molitoris, B.A.; Ronco, C.; Warnock, D.G.; Levin, A.; Acute Kidney Injury, N. Acute kidney injury network: Report of an initiative to improve outcomes in acute kidney injury. Crit. Care 2007, 11, R31. [Google Scholar] [CrossRef]
  18. Rosa, H.; Malik, A.N. Accurate measurement of cellular and cell-free circulating mitochondrial DNA content from human blood samples using real-time quantitative pcr. Methods Mol. Biol. 2021, 2277, 247–268. [Google Scholar] [CrossRef]
  19. Wallace, D.C. Mitochondrial diseases in man and mouse. Science 1999, 283, 1482–1488. [Google Scholar] [CrossRef]
  20. Song, M.J.; Lee, S.H.; Leem, A.Y.; Kim, S.Y.; Chung, K.S.; Kim, E.Y.; Jung, J.Y.; Kang, Y.A.; Kim, Y.S.; Chang, J.; et al. Predictors and outcomes of sepsis-induced cardiomyopathy in critically ill patients. Acute Crit. Care 2020, 35, 67–76. [Google Scholar] [CrossRef]
  21. Holder, A.L.; Gupta, N.; Lulaj, E.; Furgiuele, M.; Hidalgo, I.; Jones, M.P.; Jolly, T.; Gennis, P.; Birnbaum, A. Predictors of early progression to severe sepsis or shock among emergency department patients with nonsevere sepsis. Int. J. Emerg. Med. 2016, 9, 10. [Google Scholar] [CrossRef]
  22. Fry, D.E.; Pearlstein, L.; Fulton, R.L.; Polk, H.C., Jr. Multiple system organ failure. The role of uncontrolled infection. Arch. Surg. 1980, 115, 136–140. [Google Scholar] [CrossRef]
  23. Tsukamoto, T.; Chanthaphavong, R.S.; Pape, H.C. Current theories on the pathophysiology of multiple organ failure after trauma. Injury 2010, 41, 21–26. [Google Scholar] [CrossRef]
  24. Botha, A.J.; Moore, F.A.; Moore, E.E.; Kim, F.J.; Banerjee, A.; Peterson, V.M. Postinjury neutrophil priming and activation: An early vulnerable window. Surgery 1995, 118, 358–364, discussion 364–355. [Google Scholar] [CrossRef]
  25. Moore, E.E.; Moore, F.A.; Harken, A.H.; Johnson, J.L.; Ciesla, D.; Banerjee, A. The two-event construct of postinjury multiple organ failure. Shock 2005, 24 (Suppl. S1), 71–74. [Google Scholar] [CrossRef]
  26. Llompart-Pou, J.A.; Talayero, M.; Homar, J.; Royo, C.; Grupo de trabajo de Trauma y Neurointensivismo de SEMICYUC. Multiorgan failure in the serious trauma patient. Med. Intensiva 2014, 38, 455–462. [Google Scholar] [CrossRef]
  27. Venkata, C.; Kashyap, R.; Farmer, J.C.; Afessa, B. Thrombocytopenia in adult patients with sepsis: Incidence, risk factors, and its association with clinical outcome. J. Intensive Care 2013, 1, 9. [Google Scholar] [CrossRef]
  28. Vanderschueren, S.; De Weerdt, A.; Malbrain, M.; Vankersschaever, D.; Frans, E.; Wilmer, A.; Bobbaers, H. Thrombocytopenia and prognosis in intensive care. Crit. Care Med. 2000, 28, 1871–1876. [Google Scholar] [CrossRef]
  29. Akca, S.; Haji-Michael, P.; de Mendonca, A.; Suter, P.; Levi, M.; Vincent, J.L. Time course of platelet counts in critically ill patients. Crit. Care Med. 2002, 30, 753–756. [Google Scholar] [CrossRef]
  30. Terregino, C.A.; Lopez, B.L.; Karras, D.J.; Killian, A.J.; Arnold, G.K. Endogenous mediators in emergency department patients with presumed sepsis: Are levels associated with progression to severe sepsis and death? Ann. Emerg. Med. 2000, 35, 26–34. [Google Scholar] [CrossRef]
  31. Kirkman, E.; Watts, S. Haemodynamic changes in trauma. Br. J. Anaesth. 2014, 113, 266–275. [Google Scholar] [CrossRef]
  32. Collins, L.V.; Hajizadeh, S.; Holme, E.; Jonsson, I.M.; Tarkowski, A. Endogenously oxidized mitochondrial DNA induces in vivo and in vitro inflammatory responses. J. Leukoc. Biol. 2004, 75, 995–1000. [Google Scholar] [CrossRef]
  33. Riley, J.S.; Tait, S.W. Mitochondrial DNA in inflammation and immunity. EMBO Rep. 2020, 21, e49799. [Google Scholar] [CrossRef]
  34. Aarreberg, L.D.; Esser-Nobis, K.; Driscoll, C.; Shuvarikov, A.; Roby, J.A.; Gale, M., Jr. Interleukin-1beta induces mtdna release to activate innate immune signaling via cgas-sting. Mol. Cell 2019, 74, 801–815.e806. [Google Scholar] [CrossRef]
  35. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type i interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef]
  36. Harrington, J.S.; Huh, J.W.; Schenck, E.J.; Nakahira, K.; Siempos, I.I.; Choi, A.M.K. Circulating mitochondrial DNA as predictor of mortality in critically ill patients: A systematic review of clinical studies. Chest 2019, 156, 1120–1136. [Google Scholar] [CrossRef]
  37. Lan, R.; Geng, H.; Singha, P.K.; Saikumar, P.; Bottinger, E.P.; Weinberg, J.M.; Venkatachalam, M.A. Mitochondrial pathology and glycolytic shift during proximal tubule atrophy after ischemic aki. J. Am. Soc. Nephrol. 2016, 27, 3356–3367. [Google Scholar] [CrossRef]
  38. Wakabayashi, T.; Karbowski, M. Structural changes of mitochondria related to apoptosis. Biol. Signals Recept. 2001, 10, 26–56. [Google Scholar] [CrossRef]
  39. Yan, Y.; Ma, Z.; Zhu, J.; Zeng, M.; Liu, H.; Dong, Z. Mir-214 represses mitofusin-2 to promote renal tubular apoptosis in ischemic acute kidney injury. Am. J. Physiol. Renal Physiol. 2020, 318, F878–F887. [Google Scholar] [CrossRef]
  40. Zhang, X.; Agborbesong, E.; Li, X. The role of mitochondria in acute kidney injury and chronic kidney disease and its therapeutic potential. Int. J. Mol. Sci. 2021, 22, 1253. [Google Scholar] [CrossRef]
  41. Hu, Q.; Ren, J.; Wu, J.; Li, G.; Wu, X.; Liu, S.; Wang, G.; Gu, G.; Ren, H.; Hong, Z.; et al. Urinary mitochondrial DNA levels identify acute kidney injury in surgical critical illness patients. Shock 2017, 48, 11–17. [Google Scholar] [CrossRef]
  42. Whitaker, R.M.; Stallons, L.J.; Kneff, J.E.; Alge, J.L.; Harmon, J.L.; Rahn, J.J.; Arthur, J.M.; Beeson, C.C.; Chan, S.L.; Schnellmann, R.G. Urinary mitochondrial DNA is a biomarker of mitochondrial disruption and renal dysfunction in acute kidney injury. Kidney Int. 2015, 88, 1336–1344. [Google Scholar] [CrossRef]
  43. Woitas, R.P.; Stoffel-Wagner, B.; Poege, U.; Schiedermaier, P.; Spengler, U.; Sauerbruch, T. Low-molecular weight proteins as markers for glomerular filtration rate. Clin. Chem. 2001, 47, 2179–2180. [Google Scholar] [CrossRef]
  44. Filler, G.; Priem, F.; Lepage, N.; Sinha, P.; Vollmer, I.; Clark, H.; Keely, E.; Matzinger, M.; Akbari, A.; Althaus, H.; et al. Beta-trace protein, cystatin c, beta(2)-microglobulin, and creatinine compared for detecting impaired glomerular filtration rates in children. Clin. Chem. 2002, 48, 729–736. [Google Scholar] [CrossRef]
  45. Hofbauer, D.; Mougiakakos, D.; Broggini, L.; Zaiss, M.; Buttner-Herold, M.; Bach, C.; Spriewald, B.; Neumann, F.; Bisht, S.; Nolting, J.; et al. Beta(2)-microglobulin triggers nlrp3 inflammasome activation in tumor-associated macrophages to promote multiple myeloma progression. Immunity 2021, 54, 1772–1787.e9. [Google Scholar] [CrossRef]
Figure 1. (A) representative standard curve. The linear blue line represents the standard curve used for measuring standard DNAs. (B) amplification plot. The bright blue line on the far left indicates the amplification curve of standard DNA at a concentration of 1 × 108 copies/ µL, and the green line at the far right indicates the amplification curve of standard DNA at a concentration of 1 × 102 copies/µL. (C) dissociation curve for standard DNA.
Figure 1. (A) representative standard curve. The linear blue line represents the standard curve used for measuring standard DNAs. (B) amplification plot. The bright blue line on the far left indicates the amplification curve of standard DNA at a concentration of 1 × 108 copies/ µL, and the green line at the far right indicates the amplification curve of standard DNA at a concentration of 1 × 102 copies/µL. (C) dissociation curve for standard DNA.
Medicina 59 02054 g001aMedicina 59 02054 g001b
Figure 2. Receiver-operating characteristics curves for the systolic blood pressure and initial neutrophil and platelet counts between the patients with and without multiorgan distress syndrome. SBP, systolic blood pressure.
Figure 2. Receiver-operating characteristics curves for the systolic blood pressure and initial neutrophil and platelet counts between the patients with and without multiorgan distress syndrome. SBP, systolic blood pressure.
Medicina 59 02054 g002
Figure 3. Receiver-operating characteristics curves for the diastolic blood pressure and initial urine mitochondrial DNA copy number between the patients with and without acute kidney injury. DBP, diastolic blood pressure; mtDNAcn, mitochondrial DNA copy number.
Figure 3. Receiver-operating characteristics curves for the diastolic blood pressure and initial urine mitochondrial DNA copy number between the patients with and without acute kidney injury. DBP, diastolic blood pressure; mtDNAcn, mitochondrial DNA copy number.
Medicina 59 02054 g003
Table 1. Baseline characteristics in the enrolled patients.
Table 1. Baseline characteristics in the enrolled patients.
Variablesn = 83 (%)
Age63.3 ± 17.3
Male:Female56:27
Known past history
  Hypertension30 (36.1)
  Diabetes mellitus18 (21.7)
  Cerebrovascular disorder7 (8.4)
  Liver disease3 (3.6)
  Respiratory disease7 (8.4)
Diagnosis
  Cervical spine fracture1 (1.2)
  Diaphragmatic rupture1 (1.2)
  Femur fracture3 (3.6)
  Fournier’s gangrene1 (1.2)
  Hemoperitoneum16 (19.2)
  Hemopneumothorax1 (1.2)
  Ischemic enteritis3 (3.6)
  Intestinal obstruction3 (3.6)
  Panperitonitis41 (49.4)
  Popliteal artery rupture1 (1.2)
  Retroperitoneal hemorrhage3 (3.6)
  Small bowel strangulation8 (9.6)
  Thoracic spine fracture1 (1.2)
Procedure
  Conservative management with observation only4 (4.8)
  Angioembolization3 (3.6)
  Surgery76 (91.6)
Table 2. Patient characteristics between patients without and with multiorgan distress syndrome.
Table 2. Patient characteristics between patients without and with multiorgan distress syndrome.
No MODS (n = 65) (%)MODS (n = 18) (%)p-Value
Age (years)62.0 ± 18.468.0 ± 11.70.317
Male sex42 (64.6)14 (77.8)0.441
Known past history
    Hypertension23 (35.4)7 (38.9)0.841
    Diabetes mellitus14 (21.5)4 (22.2)1.000
    Cerebrovascular disorder4 (6.2)3 (16.7)0.148
    Liver disease1 (1.5)2 (11.1)0.105
    Respiratory disease6 (9.2)1 (5.6)1.000
SBP (mmHg)126.1 ± 26.598.2 ± 31.5<0.001
DBP (mmHg)70.6 ± 16.455.2 ± 15.40.001
PR (/min)93.9 ± 45.096.7 ± 28.50.133
BT (°C)36.8 ± 0.836.5 ± 1.40.205
Initial laboratory findings
    DNI (%)4.1 ± 8.49.8 ± 12.20.016
    WBC (X3) (109/L)12.8 ± 5.610.0 ± 4.90.046
    Neutrophil (X3) (109/L)10.8 ± 5.27.8 ± 3.80.028
    Creatinine (mg/dL)0.8 ± 0.31.2 ± 0.3<0.001
    Serum β2 microglobulin (mg/L)2.5 ± 2.04.2 ± 4.10.008
    Urine β2 microglobulin (mg/L)5.9 ± 11.04.6 ± 5.00.548
    Hemoglobin (g/dL)13.1 ± 2.311.8 ± 1.90.026
    Platelet (X3) (109/L)261.0 ± 107.3188.9 ± 81.60.004
    INR1.0 ± 0.11.2 ± 0.2<0.001
    CRP (mg/dL)5.7 ± 9.16.0 ± 10.10.958
    Lactate (mmol/L)2.5 ± 1.94.7 ± 3.90.005
    Serum mtDNAcn (copies/μL)716.0 ± 1357.61311.8 ± 1872.60.260
    Urine mtDNAcn (copies/μL)1067.5 ± 1575.83800.4 ± 7298.90.032
SOFA score2.0 ± 1.68.1 ± 2.0<0.001
Hospital DOS15.8 ± 10.128.7 ± 22.00.008
ICU DOS3.5 ± 3.79.1 ± 7.70.001
Mortality0 (0.0%)5 (27.8%)<0.001
MODS, multiorgan distress syndrome; SBP, systolic blood pressure; DBP, diastolic blood pressure; PR, pulse rate; BT, body temperature; DNI, delta neutrophil index; WBC, white blood cells; INR, international normalized ratio; CRP, C-reactive protein; mtDNAcn, mitochondrial DNA copy number; SOFA, sequential organ failure assessment; ICU, intensive care unit; DOS, duration of stay.
Table 3. Multivariate analysis using a logistic regression model to predict multiorgan distress syndrome.
Table 3. Multivariate analysis using a logistic regression model to predict multiorgan distress syndrome.
Odds Ratio (95% CI)p-Value
SBP (mmHg)0.97 (0.94–1.00)0.025
Initial neutrophil (X3) (109/L)0.84 (0.72–0.98)0.024
Initial creatinine (mg/dL)8.27 (0.70–97.12)0.093
Initial serum β2 microglobulin (mg/L)1.30 (1.00–1.69)0.053
Initial platelet (X3) (109/L)0.99 (0.98–1.00)0.036
CI, confidence interval; SBP, systolic blood pressure.
Table 4. Systolic blood pressure and initial neutrophil and platelet count characteristics as independent predictors of multiorgan distress syndrome.
Table 4. Systolic blood pressure and initial neutrophil and platelet count characteristics as independent predictors of multiorgan distress syndrome.
Optimal Cut-Off ValueSensitivity %Specificity %AUC (95% CI)
SBP (mmHg)11366.773.90.750 (0.606–0.877)
Initial neutrophil count (X3) (109/L)8.6572.264.60.670 (0.539–0.801)
Initial platelet count (X3) (109/L)19566.781.50.724 (0.570–0.877)
AUC, area under the curve; CI, confidence interval; SBP, systolic blood pressure.
Table 5. Patient characteristics between patients without and with acute kidney injury.
Table 5. Patient characteristics between patients without and with acute kidney injury.
No AKI (n = 44) (%)AKI (n = 36) (%)p-Value
Age (years)59.4 ± 17.966.2 ± 15.50.072
Male sex30 (68.2)24 (66.7)1.000
Known past history
    Hypertension13 (29.5)15 (41.7)0.371
    Diabetes mellitus8 (18.2)9 (25.0)0.641
    Cerebrovascular disorder3 (6.8)3 (8.3)1.000
    Liver disease2 (4.5)1 (2.8)1.000
    Respiratory disease0 (0.0)1 (2.8)1.000
SBP (mmHg)127.2 ± 19.3112.8 ± 37.90.044
DBP (mmHg)74.4 ± 13.858.6 ± 17.8<0.001
PR (/min)88.0 ± 26.2100.9 ± 56.00.183
BT (°C)36.8 ± 1.036.6 ± 0.90.276
Initial laboratory findings
    DNI (%)2.7 ± 4.87.3 ± 11.00.035
    WBC (X3) (109/L)15.1 ± 13.411.6 ± 5.90.085
    Neutrophil (X3) (109/L)11.2 ± 4.79.3 ± 5.20.045
    Creatinine (mg/dL)0.8 ± 0.21.0 ± 0.40.006
    Serum β2 microglobulin (mg/L)2.1 ± 1.43.4 ± 3.40.020
    Urine β2 microglobulin (mg/L)6.2 ± 10.65.3 ± 9.90.306
    Hemoglobin13.4 ± 2.112.2 ± 2.30.011
    Platelet (X3) (109/L)263.6 ± 93.6222.6 ± 120.60.016
    INR1.0 ± 0.11.1 ± 0.20.007
    CRP (mg/dL)5.0 ± 8.85.4 ± 9.00.815
    Lactate (mmol/L)2.2 ± 1.53.9 ± 3.30.004
    Serum mtDNAcn (copies/μL)532.6 ± 812.31251.4 ± 2027.60.158
    Urine mtDNAcn (copies/μL)561.4 ± 349.93001.8 ± 5424.50.001
SOFA score2.2 ± 2.04.5 ± 3.70.004
Hospital DOS15.9 ± 14.721.6 ± 14.30.007
ICU DOS3.1 ± 3.76.5 ± 6.40.010
MODS4 (9.1)14 (38.9)0.004
Mortality0 (0.0)5 (13.9)0.037
AKI, acute kidney injury; SBP, systolic blood pressure; DBP, diastolic blood pressure; PR, pulse rate; BT, body temperature; DNI, delta neutrophil index; WBC, white blood cells; INR, international normalized ratio; CRP, C-reactive protein; mtDNAcn, mitochondrial DNA copy number; SOFA, sequential organ failure assessment; ICU, intensive care unit; DOS, duration of stay; MODS, multiorgan distress syndrome.
Table 6. Multivariate analysis using a logistic regression model to predict acute kidney injury.
Table 6. Multivariate analysis using a logistic regression model to predict acute kidney injury.
Odds Ratio (95% CI)p-Value
SBP (mmHg)1.04 (1.00–1.07)0.059
DBP (mmHg)0.90 (0.84–0.96)0.001
Initial neutrophil (X3) (109/L)0.90 (0.79–1.03)0.127
Initial urine mtDNAcn (copies/μL)1.00 (1.00–1.00)0.010
CI, confidence interval; SBP, systolic blood pressure; DBP, diastolic blood pressure; mtDNAcn, mitochondrial DNA copy number.
Table 7. Diastolic blood pressure and initial urine mitochondrial DNA copy number as independent predictors of acute kidney injury.
Table 7. Diastolic blood pressure and initial urine mitochondrial DNA copy number as independent predictors of acute kidney injury.
Optimal Cut-Off ValueSensitivity %Specificity %AUC (95% CI)
DBP (mmHg)68.561.179.50.755 (0.647–0.864)
Initial urine mtDNAcn (copies/μL)1225.655.695.50.718 (0.590–0.845)
AUC, area under the curve; DBP, diastolic blood pressure; mtDNAcn, mitochondrial DNA copy number.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shin, I.S.; Kim, D.K.; An, S.; Gong, S.C.; Kim, M.H.; Rahman, M.H.; Kim, C.-S.; Sohn, J.H.; Kim, K.; Ryu, H. Biomarkers to Predict Multiorgan Distress Syndrome and Acute Kidney Injury in Critically Ill Surgical Patients. Medicina 2023, 59, 2054. https://doi.org/10.3390/medicina59122054

AMA Style

Shin IS, Kim DK, An S, Gong SC, Kim MH, Rahman MH, Kim C-S, Sohn JH, Kim K, Ryu H. Biomarkers to Predict Multiorgan Distress Syndrome and Acute Kidney Injury in Critically Ill Surgical Patients. Medicina. 2023; 59(12):2054. https://doi.org/10.3390/medicina59122054

Chicago/Turabian Style

Shin, In Sik, Da Kyung Kim, Sanghyun An, Sung Chan Gong, Moo Hyun Kim, Md Habibur Rahman, Cheol-Su Kim, Joon Hyeong Sohn, Kwangmin Kim, and Hoon Ryu. 2023. "Biomarkers to Predict Multiorgan Distress Syndrome and Acute Kidney Injury in Critically Ill Surgical Patients" Medicina 59, no. 12: 2054. https://doi.org/10.3390/medicina59122054

Article Metrics

Back to TopTop