Next Article in Journal
Ultrasound Evaluation of Sarcopenia in Patients with Hepatocellular Carcinoma: A Faster and Easier Way to Detect Patients at Risk
Next Article in Special Issue
Characterisation and Outcomes of Patients with Solid Organ Malignancies Admitted to the Intensive Care Unit: Mortality and Impact on Functional Status and Oncological Treatment
Previous Article in Journal
Current Trends in Diagnosis and Treatment Approach of Diabetic Retinopathy during Pregnancy: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Soluble Semaphorin 4D Serum Concentrations Are Elevated in Critically Ill Patients with Liver Cirrhosis and Correlate with Aminotransferases

1
Department of Medicine III, RWTH-University Hospital Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
2
Institute of Laboratory Medicine, Western Palatinate Hospital, 67655 Kaiserslautern, Germany
3
Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany
4
Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH-University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
*
Author to whom correspondence should be addressed.
Diagnostics 2024, 14(4), 370; https://doi.org/10.3390/diagnostics14040370
Submission received: 4 January 2024 / Revised: 29 January 2024 / Accepted: 5 February 2024 / Published: 8 February 2024
(This article belongs to the Special Issue ICU Disease Diagnosis)

Abstract

:
Semaphorin 4D (Sema4D), also known as CD100, is a multifunctional transmembrane protein with immunoregulatory functions. Upon the activation of immune cells, soluble Semaphorin 4D (sSema4D) is proteolytically cleaved from the membrane by metalloproteinases. sSema4D levels are elevated in various (auto-)inflammatory diseases. Our aim was to investigate sSema4D levels in association with sepsis and critical illnesses and to evaluate sSema4D’s potential as a prognostic biomarker. We measured sSema4D levels in 192 patients upon admission to our medical intensive care unit. We found similar levels of sSema4D in 125 patients with sepsis compared to 67 non-septic patients. sSema4D levels correlated with leukocytes but not with other markers of systemic inflammation such as C-reactive protein or procalcitonin. Most interestingly, in a subgroup of patients suffering from pre-existing liver cirrhosis, we observed significantly higher levels of sSema4D. Consistently, sSema4D was also positively correlated with markers of hepatic and cholestatic injury. Our study suggests that sSema4D is not regulated in sepsis compared to other causes of critical illness. However, sSema4D seems to be associated with hepatic injury and inflammation.

1. Introduction

Semaphorin 4D (Sema4D), also known as CD100, is a multifunctional 150 kDa transmembrane protein expressed in various tissues, such as endothelial cells, platelets, and immune cells including T-cells, B-cells, neutrophils and monocytes. Upon cell activation, soluble Semaphorin 4D (sSema4D) or soluble CD100 (sCD100), a 120 kDa protein, is proteolytically cleaved from the membrane by metalloproteinases to form a soluble homodimer, which retains the same functions as its membrane-bound form [1,2,3,4]. As part of the semaphore family, Sema4D is a key player in axon guidance during neural development, mainly in a repulsive manner [5,6]. In recent years, Sema4D has gained attention for its role in immune response and autoimmunity and has been investigated as a prognostic biomarker and potential therapeutic target. There is currently a single-center observational study ongoing that will compare blood concentrations of Semaphorins 1 to 7 in patients with early septic shock to those of postoperative patients suffering systemic inflammatory response syndrome (clinicaltrials.gov, NCT02692118) [7].
Sema4D primarily exerts its effects through receptors of the plexin family, namely Plexin B1, B2 and C1, which are found in many tissues and cells except the immune system. These cells control axon-guidance processes and endothelial cell migration [6,8,9]. Among the plexin family, Plexin B1 is the main receptor with the highest affinity for Sema4D [10,11].
CD72 is the main receptor for Sema4D in the immune system, although it binds to Sema4D with a much lower affinity than plexins [11,12]. CD72 is expressed in lymph nodes and the spleen as well as B cells, dendritic cells and natural killer cells, regulating their activation, proliferation and migration [12,13,14,15,16,17].
During coagulation, Sema4D performs a dual role. At the beginning of thrombus formation, Sema4D binds to the receptors of nearby platelets, which promotes thrombus formation. This is followed by the shedding of Sema4D from the cell surface as the thrombus grows. This allows sSema4D to interact with other cell types such as endothelial cells and monocytes [4].
In autoimmune disease, sSema4D has been described as pro-inflammatory and was found to correlate with disease activity, making it a promising biomarker in this context. So far, it has best been characterized in patients with rheumatoid arthritis (RA), patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) and children with Kawasaki disease [18,19,20].
In patients with rheumatoid arthritis, serum sSema4D levels were shown to be significantly higher than in healthy controls; however, in patients with osteoarthritis, ankylosing spondylitis and systemic lupus erythematodes, sSema4D levels were not increased compared to healthy controls. In patients with RA, serum levels of sSema4D were positively correlated with disease activity markers, C-reactive protein (CRP) levels, rheumatoid factor titers and bone metabolic markers. Yoshida et al. demonstrated a significant decrease in sSema4D levels after 6 months in patients who responded to a biological DMARD therapy. This decrease was not observed in patients who only moderately responded or did not respond at all to a 6-month course of DMARD therapy [18]. The increase in sSema4D serum levels was accompanied by a decrease in cell surface expression especially on CD3+ and CD14+ cells, suggesting that the elevation of sSema4D resulted from increased shedding from membrane-bound Sema4D, particularly by ADAMTS-4. Treatment of CD14+ monocytes with sSema4D induced the production of the proinflammatory cytokines IL-6 and TNF-α.
Patients with AAV have significantly higher serum levels of sSema4D compared to patients with bacterial infections and healthy controls. Furthermore, sSema4D levels are positively correlated with vasculitis activity scores and neutrophil counts, but no correlation with CRP or MPO-ANCA titers has been shown. In addition, Nishide et al. showed that the cell surface expression of Sema4D in polymorphonuclear cells was reduced in patients with AAV compared to healthy controls, which was attributed to the shedding of Sema4D from activated neutrophils, primarily mediated via the metalloproteinase ADAMTS-17 [19].
Wang et al. recently showed that Sema4D is highly expressed in liver samples from patients with fibrotic liver disease compared to healthy controls. The main sources of Sema4D in fibrotic liver are activated hepatic stellate cells followed by hepatocytes and endothelial cells. Using different mouse models, they demonstrated that Sema4D knockout suppresses liver fibrosis, which is partly mediated by regulating the balance of Th1, Th2, Th17 and T-bet positive Treg cells [21].
Given its complex function regarding immune response and immunological cell migration, we hypothesized that sSema4D may play an important role in the setting of critically ill and septic patients and serve as a biomarker for the prediction of disease severity or survival. So far, sSema4D has not been investigated in this setting. Therefore, we conducted a study in our tertiary care medical intensive care unit (ICU) with a cohort of septic and non-septic patients to elucidate sSema4D’s clinical relevance and potential prognostic value.

2. Materials and Methods

2.1. Selection and Inclusion of Patients

In this observational cohort study, we enrolled 192 patients, in a prospective manner, who were consecutively admitted to our tertiary care medical intensive care unit at the Department of Gastroenterology, Digestive Diseases, and Intensive Care Medicine of University Hospital RWTH Aachen, Germany, from 2006 to 2012. Patients above 18 years of age and with written informed consent for inclusion in the study (from the patient, their spouse or appointed legal guardian) were considered for inclusion. Patients with an expected short-term ICU stay (<48 h), patients admitted from another ICU and patients with poisoning were excluded, as previously described [22,23,24]. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) were used to differentiate between septic and non-septic patients, and treatment was conducted according to guidelines [25]. Our study was approved by our local ethics committee and conducted in accordance with the ethical standards stated in the Declaration of Helsinki (reference number EK150/06). For the assessment of long-term survival, the patient, their spouse or primary care physician were contacted at 6-month intervals for 2 years after discharge from the ICU.

2.2. Laboratory Analysis of Circulating Soluble Semaphorin4D in Patient Serum

Blood samples were collected from patients upon admission to the ICU. Following centrifugation for 10 min at 4 °C, serum was aliquoted and stored at −80 °C until further analysis. The quantitative sandwich enzyme-linked immunosorbent assay (ELISA) for soluble Semaphorin 4D (Soluble Semaphorin 4D ELISA kit, BI-20405, Biomedica Medizinprodukte GmbH, Vienna, Austria) was performed according to the manufacturer’s instructions [26]. Measurements of sSema4D were performed without knowledge of any clinical or laboratory data from the patients.

2.3. Statistical Analysis

Statistical analyses were performed and graphs were created using SPSS version 29 (SPSS, Chicago, IL, USA) and the packages NumPy version 1.21.5 [27], Pandas version 1.4.4 [28], Matplotlib version 3.5.2 [29], Seaborn version 0.11.2 [30], Pingouin version 0.5.3 [31], Scikit-learn version 1.0.2 [32] and Lifelines [33] in Jupyter Notebooks version 6.5.4 [34] using Python version 3.11 [35]. Due to the skewed distribution of most parameters, data are presented as the median and range. As a normal distribution could not be assumed, either the two-tailed Mann–Whitney U test or the Chi-squared test was applied to assess differences between groups. In cases with more than two groups, the Kruskal–Wallis test was applied. Statistical significance was assumed where p < 0.05 for all calculations. Spearman’s rank correlation test was used to analyze the associations between sSema4D and the other variables. The Youden Index (the sum of sensitivity and specificity minus one) was calculated and then used as the optimal cut-off value for the prognosis analyses. A receiver operating characteristic (ROC) curve with the corresponding area under the curve (AUC) was determined to evaluate the quality of the prognostic marker. Kaplan–Meier curve analysis, including 95% confidence intervals, was applied to examine differences in survival. A log-rank test was used to assess significance.

3. Results

3.1. Soluble Semaphorin 4D Serum Concentrations Do Not Differ between Septic and Non-Septic Critically Ill Patients

A total of 192 patients were included in this study. Of these, 125 were admitted to the ICU due to sepsis and 67 were admitted due to other critical illness. The median age of the study cohort was 64.5 years, ranging from 18 to 89, and there was no significant difference between septic and non-septic patients. Of these patients, 113 (58.9%) were male and 79 (41.1%) were female. There were no differences in age, gender distribution, comorbidities (measured using the Charlson Comorbidity Index) or mortality between the two groups.
We did not observe any difference in sSema4D concentrations between septic and non-septic critically ill patients (Figure 1).
As expected, septic patients more frequently required mechanical ventilation (73.6% vs. 57.5%, p = 0.036) as well as vasopressor therapy (70% vs. 47.4%, p = 0.005). Disease severity, as assessed using the APACHE II score, was higher in septic patients (median of 18 vs. 16 points, p = 0.039) and organ failure, as assessed using the SOFA score, was more severe in sepsis patients (median of 11 vs. 7 points, p = 0.006). Consistent with this, septic patients required longer ICU treatment than non-septic patients (median of 10 vs. 6 days, p < 0.001; Table 1).

3.2. Soluble Semaphorin 4D Levels Are Not Associated with Disease Etiology

In our cohort, the most common type of infection in sepsis patients was pulmonary (55.2%). Other origins of sepsis were abdominal (15.2%) or urogenital (8%). The remaining sepsis patients (21.6%) were treated due to blood-stream infections, skin infections or an unknown site of infection. We observed a trend towards higher sSema4D levels in sepsis patients with other sites of infection, with a median of 44.62 ng/mL (range: 15.70–184.60 ng/mL) compared to patients with a pulmonary focus (median: 33.11 ng/mL; range 9.99–240.00 ng/mL), an abdominal focus (median: 36.69 ng/mL; range: 11.12–160.27 ng/mL) or a urogenital focus (median: 34.51 ng/mL; range 16.30–82.13 ng/mL). However, these differences were not statistically significant (p = 0.059) (Table 2).
The non-septic critically ill patients were admitted to our ICU due to cardiocirculatory disorders (19.4%), advanced liver disease (19.4%), respiratory failure (14.9%) and a variety of other diseases (46.3%). In this group, the regulation of sSema4D levels was not observed (p = 0.809) (Table 2).

3.3. Soluble Semaphorin 4D Serum Concentrations Are Increased in Patients with Liver Cirrhosis and Positively Correlated with Liver Function Tests

We set out to test whether certain comorbidities had an influence on sSema4D concentrations. Patients with a history of diabetes, coronary artery disease, hypertension, chronic obstructive lung disease, chronic alcohol abuse and active malignancy did not have altered sSema4D concentrations (Table 3).
However, patients with pre-existing liver cirrhosis (n = 20) showed significantly higher sSema4D levels, with a median of 47.25 ng/mL (range: 15.14–163.01 ng/mL, p = 0.02). Etiologies of liver cirrhosis were alcohol related (n = 8) or due to chronic hepatitis B (n = 2), chronic hepatitis C (n = 2), primary biliary cholangitis (n = 2), non-alcoholic steatohepatitis (n = 1), cardial dysfunction (n = 1) or cryptogenic (n = 4). Moreover, sSema4D was positively correlated with liver function tests. We observed a weak correlation between sSema4D and alanine aminotransferase (ALT, Spearman’s r = 0.160, p = 0.027) and a correlation of moderate strength with aspartate aminotransferase (AST, Spearman’s r = 0.258, p < 0.001) as markers of hepatic injury and inflammation. The cholestatic parameters bilirubin (Spearman’s r = 0.170, p = 0.019) and gamma-glutamyl transpeptidase (γGT, Spearman’s r = 0.237, p = 0.001) also showed weak and moderate correlations with sSema4D, respectively (Table 4).

3.4. Soluble Semaphorin 4D Serum Concentrations Correlate with Leukocyte Count but Not with Other Markers of Systemic Inflammation

To identify the factors that influence sSema4D levels, we performed extensive correlation analyses between sSema4D and different markers of inflammation, laboratory values, clinical scores and demographics. sSema4D correlated with leukocyte count (Spearman’s r = 0.144, p = 0.048) but not with other established markers of inflammation such as C-reactive protein, procalcitonin and interleukin-6 or interleukin-10. We observed a weak correlation between sSema4D and triglycerides (Spearman’s r = 0.201, p = 0.012) while other metabolic markers, such as cholesterol, high-density lipoprotein, low-density lipoprotein, glycosylated hemoglobin A1, blood glucose, insulin and C-peptide, showed no correlation. Body mass index, age, markers of renal function and plasmatic coagulation markers also showed no correlation. Underlining the fact that sSema4D concentrations did not differ between septic and non-septic patients, we found no correlation with clinical markers of disease severity (APACHE-II, SOFA, Horovitz, vasopressor demand and FiO2 demand) (Table 4).

3.5. Soluble Semaphorin 4D Serum Concentrations Do Not Predict Short-Term or Long-Term Mortality

To examine the potential prognostic value of sSema4D, we compared the baseline sSema4D serum levels of surviving and deceased patients at different timepoints.
There was no significant difference in sSema4D at the baseline between the groups at 30 days after admission (p = 0.343; Figure 2A), 60 days after admission (p = 0.243; Figure 2B), 90 days after admission (p = 0.381; Figure 2C), 180 days after admission (p = 0.755; Figure 2D) or a year after admission (p = 0.630; Figure 2E) to the ICU. However, at all the timepoints mentioned above, baseline median sSema4D levels were lower in surviving patients than in deceased patients. We performed a receiver operating characteristic (ROC) curve analysis in which sSema4D levels showed an area under the curve (AUC) of 0.524 for the prediction of survival after one year (Figure 3A). In addition, we conducted Kaplan–Meier curve analyses using the Youden Index to establish the ideal cut-off value for the prediction of survival at the one-year observation period for all patients. This showed a trend towards improved survival for patients with baseline sSema4D < 41.74 ng/mL (log-rank 4.045, p = 0.044; Figure 3B). Although the log-rank test, which compares the median survival times between groups, suggests a significant difference, the 95% confidence intervals overlap during the whole observation period of one year. Potentially, a significant difference in survival is reached after one year.

4. Discussion

Sema4D has gained recent attention due to its diverse functions in the immune system; it is involved in T-cell-priming, antibody production and cell-to-cell interactions. Therefore, it has been studied in the context of inflammatory diseases, viral infections, angiogenesis and cancer as both a prognostic biomarker and a potential therapeutic target [2].
Different associations with sSema4D levels have been previously described in infectious diseases. In patients with hemorrhagic fever with renal syndrome (HFRS) caused by the Hantaan virus, sSema4D levels were significantly higher in the acute phase compared to healthy controls [36]. Higher levels of sSema4D were associated with disease severity as wells as corresponding clinical parameters. In the convalescent phase, the sSema4D levels of patients with HFRS declined but still remained higher than in healthy controls. In patients with acute hepatitis C virus infection increased serum sSema4D levels have been observed, which dropped to the level of healthy controls after sustained virological response [37]. However, in patients with chronic HIV, sSema4D levels are decreased compared to healthy controls before and 2 years after successful highly-active anti-retroviral therapy regardless of the CD4 and CD8 cell counts [38].
In our study, we did not observe a difference in sSema4D between septic and non-septic critically ill patients. There was also no association between sSema4D levels and disease etiology, disease severity or survival. We did observe a positive correlation with leukocyte count, but not with other established markers of inflammation like CRP and PCT. Similarly, Nishide et al. found a correlation between sSema4D and neutrophil count but not with CRP in patients with AAV [19].
These findings suggest that sSema4D is not regulated in sepsis and should not be considered as a general pro-inflammatory driver in that setting. Instead, sSema4D appears to be a protein which is involved in the pathogenesis of specific diseases only. Similarly, Yoshida et al. found elevated sSema4D in the serum of patients with rheumatoid arthritis, while patients with osteoarthritis, ankylosing spondylitis and systemic lupus erythematodes had no increase in sSema4D levels compared to healthy controls [18]. Consistent with these findings, sSema4D levels in RA were independent of age and gender in our cohort.
In our study, a subgroup of patients with a history of liver cirrhosis had significantly higher levels of sSema4D. In addition, sSema4D was positively correlated with aminotransferases, as markers of hepatic inflammation, and the cholestatic liver parameters bilirubin and gamma-glutamyl transpeptidase. The majority of our patients suffered from sepsis, which is associated with hyperinflammation in the early phase. This inflammation can trigger profibrotic processes in the liver. Sema4D has recently been found to be highly expressed in human fibrotic livers and in mouse models of liver fibrosis [21]. The main source of upregulated Sema4D is activated hepatic stem cells. Knockout of Sema4D alleviated liver fibrosis in different mouse models compared to the wild-type; however, Wang et al. did not measure soluble Semaphorin 4D in patients with liver fibrosis or in mice, respectively. Nevertheless, it is reasonable to assume that increased expression of Sema4D also leads to increased shedding and elevated peripheral levels of soluble Semaphorin 4D.
We acknowledge some limitations of our study. Given its design as a single-center study, high reproducibility and technical accuracy were achieved; however, transferability of the results into other settings is limited. Our cohort was heterogeneous, including patients with various disease etiologies, and, therefore, subgroups might be underpowered to show significant changes in sSema4D levels. Moreover, we did not measure membrane-bound Semaphorin 4D on peripheral blood cells, making it difficult to understand how the up- or downregulation of sSema4D is associated with increases or decreases in shedding from, for example, leukocytes in the setting of critical illness. We also did not measure sSema4D levels at different timepoints during the course of our patients’ ICU stays, so we cannot make assumptions about changes in sSema4D levels in the later stages of critical illness and sepsis.
Another potential limitation of our study is the fact that we measured sSema4D in serum instead of plasma. We decided to do so to ensure better comparability with previous studies which had examined sSema4D serum levels in infectious diseases [38] and several autoimmune disorders [18,19,20]. Of note, sSema4D is involved in thrombus formation, during which it is shed from the cell surface of platelets. Thus, pre-analytical blood sampling procedures, which initiate coagulation processes, result in increased levels of sSema4D in serum compared to plasma levels, which further challenges interpretation [26,39].

5. Conclusions

Our study is the first to examine the potential of sSema4D as a biomarker in the setting of critical illness and sepsis. We found that sSema4D levels are comparable between septic and non-septic patients with critical illnesses. Soluble Semaphorin 4D had no prognostic value in our cohort. However, in a subgroup of patients with a history of liver cirrhosis, we observed significantly higher sSema4D levels. Furthermore, there was a positive correlation between sSema4D and aminotransferases, bilirubin and γGT.

Author Contributions

Conceptualization, S.A.J., V.K., F.T. and A.K.; data curation, S.A.J., V.K., P.H., J.K.A., L.B., M.R.P., J.F.B. and T.H.W.; formal analysis, S.A.J., V.K., P.H. and A.K.; methodology, S.A.J., P.H., E.Y., R.W. and A.K.; supervision, J.F.B., K.H., T.H.W., R.W., E.Y., C.T., F.T. and A.K.; writing—original draft, S.A.J., V.K. and A.K.; writing—review and editing, S.A.J., P.H., V.K., M.R.P., J.K.A., K.H., T.H.W., R.W., F.T. and A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the German Research Foundation (DFG; CRC1382, Project-ID 403224013). T.H.W. and R.W. were supported by grants from the Interdisciplinary Centre for Clinical Research within the Faculty of Medicine at RWTH Aachen University (grants PTD1-5 and PTD1-10).

Institutional Review Board Statement

The local ethics committee (Ethikkommission der Medizinischen Fakultät der RWTH Aachen), in accordance with the ethical standards of the Declaration of Helsinki (reference number EK 150/06), approved our study in 2 November 2006.

Informed Consent Statement

Written informed consent was obtained from each patient, his/her spouse or legal guardian.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We cordially thank Gabriela Berg (Biomedica, Austria) for excellent scientific cooperation and technical advice regarding concept development and ELISA kits.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bougeret, C.; Mansur, I.G.; Dastot, H.; Schmid, M.; Mahouy, G.; Bensussan, A.; Boumsell, L. Increased surface expression of a newly identified 150-kDa dimer early after human T lymphocyte activation. J. Immunol. 1992, 148, 318–323. [Google Scholar] [CrossRef]
  2. Maleki, K.T.; Cornillet, M.; Björkström, N.K. Soluble SEMA4D/CD100: A novel immunoregulator in infectious and inflammatory diseases. Clin. Immunol. 2016, 163, 52–59. [Google Scholar] [CrossRef]
  3. Wang, X.; Kumanogoh, A.; Watanabe, C.; Shi, W.; Yoshida, K.; Kikutani, H. Functional soluble CD100/Sema4D released from activated lymphocytes: Possible role in normal and pathologic immune responses. Blood 2001, 97, 3498–3504. [Google Scholar] [CrossRef]
  4. Zhu, L.; Bergmeier, W.; Wu, J.; Jiang, H.; Stalker, T.J.; Cieslak, M.; Fan, R.; Boumsell, L.; Kumanogoh, A.; Kikutani, H.; et al. Regulated surface expression and shedding support a dual role for semaphorin 4D in platelet responses to vascular injury. Proc. Natl. Acad. Sci. USA 2007, 104, 1621–1626. [Google Scholar] [CrossRef] [PubMed]
  5. Kolodkin, A.L. Semaphorins: Mediators of repulsive growth cone guidance. Trends Cell Biol. 1996, 6, 15–22. [Google Scholar] [CrossRef] [PubMed]
  6. Ito, Y.; Oinuma, I.; Katoh, H.; Kaibuchi, K.; Negishi, M. Sema4D/plexin-B1 activates GSK-3beta through R-Ras GAP activity, inducing growth cone collapse. EMBO Rep. 2006, 7, 704–709. [Google Scholar] [CrossRef] [PubMed]
  7. Thomas, R.; Yang, X. Semaphorins in immune cell function, inflammatory and infectious diseases. Curr. Res. Immunol. 2023, 4, 100060. [Google Scholar] [CrossRef] [PubMed]
  8. Swiercz, J.M.; Kuner, R.; Behrens, J.; Offermanns, S. Plexin-B1 directly interacts with PDZ-RhoGEF/LARG to regulate RhoA and growth cone morphology. Neuron 2002, 35, 51–63. [Google Scholar] [CrossRef] [PubMed]
  9. Conrotto, P.; Valdembri, D.; Corso, S.; Serini, G.; Tamagnone, L.; Comoglio, P.M.; Bussolino, F.; Giordano, S. Sema4D induces angiogenesis through Met recruitment by Plexin B1. Blood 2005, 105, 4321–4329. [Google Scholar] [CrossRef]
  10. Delaire, S.; Billard, C.; Tordjman, R.; Chédotal, A.; Elhabazi, A.; Bensussan, A.; Boumsell, L. Biological activity of soluble CD100. II. Soluble CD100, similarly to H-SemaIII, inhibits immune cell migration. J. Immunol. 2001, 166, 4348–4354. [Google Scholar] [CrossRef]
  11. Tamagnone, L.; Artigiani, S.; Chen, H.; He, Z.; Ming, G.I.; Song, H.; Chedotal, A.; Winberg, M.L.; Goodman, C.S.; Poo, M.; et al. Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell 1999, 99, 71–80. [Google Scholar] [CrossRef]
  12. Kumanogoh, A.; Watanabe, C.; Lee, I.; Wang, X.; Shi, W.; Araki, H.; Hirata, H.; Iwahori, K.; Uchida, J.; Yasui, T.; et al. Identification of CD72 as a lymphocyte receptor for the class IV semaphorin CD100: A novel mechanism for regulating B cell signaling. Immunity 2000, 13, 621–631. [Google Scholar] [CrossRef]
  13. Fagerberg, L.; Hallström, B.M.; Oksvold, P.; Kampf, C.; Djureinovic, D.; Odeberg, J.; Habuka, M.; Tahmasebpoor, S.; Danielsson, A.; Edlund, K.; et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol. Cell. Proteom. 2014, 13, 397–406. [Google Scholar] [CrossRef] [PubMed]
  14. Suzuki, K.; Kumanogoh, A.; Kikutani, H. Semaphorins and their receptors in immune cell interactions. Nat. Immunol. 2008, 9, 17–23. [Google Scholar] [CrossRef]
  15. Wu, H.J.; Bondada, S. CD72, a coreceptor with both positive and negative effects on B lymphocyte development and function. J. Clin. Immunol. 2009, 29, 12–21. [Google Scholar] [CrossRef]
  16. Alcón, V.L.; Luther, C.; Balce, D.; Takei, F. B-cell co-receptor CD72 is expressed on NK cells and inhibits IFN-gamma production but not cytotoxicity. Eur. J. Immunol. 2009, 39, 826–832. [Google Scholar] [CrossRef]
  17. Vadasz, Z.; Haj, T.; Balbir, A.; Peri, R.; Rosner, I.; Slobodin, G.; Kessel, A.; Toubi, E. A regulatory role for CD72 expression on B cells in systemic lupus erythematosus. Semin. Arthritis Rheum. 2014, 43, 767–771. [Google Scholar] [CrossRef] [PubMed]
  18. Yoshida, Y.; Ogata, A.; Kang, S.; Ebina, K.; Shi, K.; Nojima, S.; Kimura, T.; Ito, D.; Morimoto, K.; Nishide, M.; et al. Semaphorin 4D Contributes to Rheumatoid Arthritis by Inducing Inflammatory Cytokine Production: Pathogenic and Therapeutic Implications. Arthritis Rheumatol. 2015, 67, 1481–1490. [Google Scholar] [CrossRef]
  19. Nishide, M.; Nojima, S.; Ito, D.; Takamatsu, H.; Koyama, S.; Kang, S.; Kimura, T.; Morimoto, K.; Hosokawa, T.; Hayama, Y.; et al. Semaphorin 4D inhibits neutrophil activation and is involved in the pathogenesis of neutrophil-mediated autoimmune vasculitis. Ann. Rheum. Dis. 2017, 76, 1440–1448. [Google Scholar] [CrossRef] [PubMed]
  20. Huang, J.; Wu, S.; Cao, S.; Zhu, X.; Zhang, S. Neutrophil-Derived Semaphorin 4D Induces Inflammatory Cytokine Production of Endothelial Cells via Different Plexin Receptors in Kawasaki Disease. Biomed. Res. Int. 2020, 2020, 6663291. [Google Scholar] [CrossRef]
  21. Wang, L.; Li, D.; Zhu, Z.; Liao, Y.; Wu, J.; Liu, Y.; Yang, R.; Dai, H.; Wu, Z.; Sun, X. Knockout of Sema4D alleviates liver fibrosis by suppressing AOX1 expression. Pharmacol. Res. 2023, 195, 106886. [Google Scholar] [CrossRef]
  22. Hohlstein, P.; Brozat, J.F.; Schuler, J.; Abu Jhaisha, S.; Pollmanns, M.R.; Bündgens, L.; Wirtz, T.H.; Yagmur, E.; Hamesch, K.; Weiskirchen, R.; et al. Secreted Frizzled Related Protein 5 (SFRP5) Serum Levels Are Decreased in Critical Illness and Sepsis and Are Associated with Short-Term Mortality. Biomedicines 2023, 11, 313. [Google Scholar] [CrossRef]
  23. Yagmur, E.; Abu Jhaisha, S.; Buendgens, L.; Sapundzhieva, N.; Brozat, J.F.; Hohlstein, P.; Pollmanns, M.R.; Koek, G.H.; Weiskirchen, R.; Trautwein, C.; et al. Clusterin Plasma Concentrations Are Decreased in Sepsis and Inversely Correlated with Established Markers of Inflammation. Diagnostics 2022, 12, 3010. [Google Scholar] [CrossRef]
  24. Hohlstein, P.; Abu Jhaisha, S.; Yagmur, E.; Wawer, D.; Pollmanns, M.R.; Adams, J.K.; Wirtz, T.H.; Brozat, J.F.; Bündgens, L.; Hamesch, K.; et al. Elevated Midkine Serum Levels Are Associated with Long-Term Survival in Critically Ill Patients. Int. J. Mol. Sci. 2024, 25, 454. [Google Scholar] [CrossRef]
  25. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef]
  26. Laber, A.; Gadermaier, E.; Wallwitz, J.; Berg, G.; Himmler, G. A high-sensitivity enzyme immunoassay for the quantification of soluble human semaphorin 4D in plasma. Anal. Biochem. 2019, 574, 15–22. [Google Scholar] [CrossRef]
  27. Harris, C.R.; Millman, K.J.; van der Walt, S.J.; Gommers, R.; Virtanen, P.; Cournapeau, D.; Wieser, E.; Taylor, J.; Berg, S.; Smith, N.J.; et al. Array programming with NumPy. Nature 2020, 585, 357–362. [Google Scholar] [CrossRef] [PubMed]
  28. McKinney, W. Data Structures for Statistical Computing in Python. In Proceedings of the 9th Python in Science Conference (SciPy 2010), Austin, TX, USA, 28 June–3 July 2010; pp. 56–61. [Google Scholar]
  29. Hunter, J.D. Matplotlib: A 2D Graphics Environment. Comput. Sci. Eng. 2007, 9, 90–95. [Google Scholar] [CrossRef]
  30. Waskom, M.; Botvinnik, O.; O’Kane, D.; Hobson, P.; Lukauskas, S.; Gemperline, D.C.; Augspurger, T.; Halchenko, Y.; Cole, J.B.; Warmenhoven, J.; et al. Mwaskom/Seaborn: v0.8.1 (September 2017); European Organization for Nuclear Research: Geneva, Switzerland, 2017. [Google Scholar]
  31. Vallat, R. Pingouin: Statistics in Python. J. Open Source Softw. 2018, 3, 1026. [Google Scholar] [CrossRef]
  32. Pedregosa, F.; Varoquaux, G.; Gramfort, A.; Michel, V.; Thirion, B.; Grisel, O.; Blondel, M.; Müller, A.; Nothman, J.; Louppe, G.; et al. Scikit-learn: Machine Learning in Python. J. Mach. Learn. Res. 2011, 12, 2825–2830. [Google Scholar] [CrossRef]
  33. Davidson-Pilon, C. lifelines: Survival analysis in Python. J. Open Source Softw. 2019, 4, 1317. [Google Scholar] [CrossRef]
  34. Kluyver, T.; Ragan-Kelley, B.; Pérez, F.; Granger, B.; Bussonnier, M.; Frederic, J.; Kelley, K.; Hamrick, J.; Grout, J.; Corlay, S.; et al. Jupyter Notebooks—A publishing format for reproducible computational workflows. In Proceedings of the 20th International Conference on Electronic Publishing, Göttingen, Germany, 7–9 June 2016; pp. 87–90. [Google Scholar]
  35. Van Rossum, G.; Drake, F.L. Python 3 Reference Manual; CreateSpace: Scotts Valley, CA, USA, 2009. [Google Scholar]
  36. Liu, B.; Ma, Y.; Yi, J.; Xu, Z.; Zhang, Y.S.; Zhang, C.; Zhuang, R.; Yu, H.; Wang, J.; Yang, A.; et al. Elevated plasma soluble Sema4D/CD100 levels are associated with disease severity in patients of hemorrhagic fever with renal syndrome. PLoS ONE 2013, 8, e73958. [Google Scholar] [CrossRef]
  37. He, Y.; Li, B.; Zhou, Y.; Zhang, Y.; Guo, Y.; Ji, G.; Fan, C.; Jia, Z. Alteration of CD100 expression on natural killer cells in chronic patients with hepatitis C virus before and after initiation of antiviral treatment. Xi Bao Yu Fen. Zi Mian Yi Xue Za Zhi 2014, 30, 856–860. [Google Scholar]
  38. Vadasz, Z.; Elbirt, D.; Radian, S.; Bezalel-Rosenberg, S.; Mahlab-Guri, K.; Toubi, E.; Asher, I.; Sthoeger, Z. Low levels of the immunoregulator Semaphorin 4D (CD100) in sera of HIV patients. Clin. Immunol. 2018, 191, 88–93. [Google Scholar] [CrossRef] [PubMed]
  39. Liu, Z.; Zhang, C.; Li, Q.; Xiao, L.; Kong, X.; Xu, X.; Shi, B. Effects of different processing methods and durations of blood specimens on the level of soluble CD100 in peripheral blood. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2015, 31, 1375–1377, 1382. [Google Scholar] [PubMed]
Figure 1. Serum sSema4D concentrations in critically ill patients with and without sepsis (A) and comparison between the sexes (B). Sample sizes: patients n = 192, non-sepsis n = 67, sepsis n = 125. Significance of the difference between groups was assessed using the Mann-Whitney U test. p-values < 0.05 were considered statistically significant.
Figure 1. Serum sSema4D concentrations in critically ill patients with and without sepsis (A) and comparison between the sexes (B). Sample sizes: patients n = 192, non-sepsis n = 67, sepsis n = 125. Significance of the difference between groups was assessed using the Mann-Whitney U test. p-values < 0.05 were considered statistically significant.
Diagnostics 14 00370 g001
Figure 2. sSema4D levels in a consecutive survival analysis of critically ill patients treated in the ICU. (AE) Survival status at days 30 through 365. The sample size was n = 192 patients. Significance of the difference between groups was assessed using the Mann–Whitney U test. p-values < 0.05 were considered statistically significant.
Figure 2. sSema4D levels in a consecutive survival analysis of critically ill patients treated in the ICU. (AE) Survival status at days 30 through 365. The sample size was n = 192 patients. Significance of the difference between groups was assessed using the Mann–Whitney U test. p-values < 0.05 were considered statistically significant.
Diagnostics 14 00370 g002
Figure 3. Receiver operating characteristic (ROC) curves for the prediction of one-year survival using serum sSema4D levels in all patients (A). Kaplan–Meier curves for sSema4D < 41.74 ng/mL (orange) and ≥41.74 ng/mL (blue) in all patients (B). Censored events are indicated by a crossing vertical line. Cut-off values of the Kaplan–Meier curve were determined using the Youden Index for all patients. Sample size: patients n = 192. Significance of the difference between groups was assessed using the log-rank test. p-values < 0.05 were considered statistically significant and are denoted by an asterisk (“*”). Abbreviations: AUC: area under curve. Shaded areas in the Kaplan–Meier curves represent the 95% confidence intervals.
Figure 3. Receiver operating characteristic (ROC) curves for the prediction of one-year survival using serum sSema4D levels in all patients (A). Kaplan–Meier curves for sSema4D < 41.74 ng/mL (orange) and ≥41.74 ng/mL (blue) in all patients (B). Censored events are indicated by a crossing vertical line. Cut-off values of the Kaplan–Meier curve were determined using the Youden Index for all patients. Sample size: patients n = 192. Significance of the difference between groups was assessed using the log-rank test. p-values < 0.05 were considered statistically significant and are denoted by an asterisk (“*”). Abbreviations: AUC: area under curve. Shaded areas in the Kaplan–Meier curves represent the 95% confidence intervals.
Diagnostics 14 00370 g003
Table 1. Baseline patient characteristics.
Table 1. Baseline patient characteristics.
ParameterAll PatientsSepsisNon-Sepsisp
Number n19212567
Sex (male/female) n113/7978/4735/320.226
Age (years)64.5 (18–89)65 (21–89)63 (18–87)0.663
APACHE II score17 (2–40)18 (3–40)16 (2–37)0.039 *
SOFA score10 (0–18)11 (3–17)7 (0–18)0.006 *
Charlson Comorbidity index4 (0–16)4 (0–16)4 (0–13)0.297
Mechanical ventilation n (%)130 (68.0)92 (73.6)38 (57.5)0.036 *
Vasopressor demand n (%)115 (62.2)84 (70.0)31 (47.7)0.005 *
ICU days n8 (1–137)10 (1–137)6 (2–44)<0.001 *
Death in ICU n (%)52 (27.1)38 (30.4)14 (20.9)0.214
30-day mortality n (%)57 (34.5)41 (36.3)16 (30.8)0.606
1-year mortality n (%)88 (59.9)65 (64.4)23 (50.0)0.142
sSema4D (ng/mL)35.98 (9.99–240.00)35.38 (9.99–240.00)36.83 (12.26–203.30)0.522
The median and range (in parentheses) are given unless indicated otherwise. Abbreviations: APACHE: Acute Physiology and Chronic Health Evaluation; SOFA: Sepsis-Related Organ Failure Assessment; ICU: intensive care unit. Significance of the difference between sepsis and non-sepsis patients was assessed using the Mann–Whitney U test or the Chi-squared test, respectively. p-values < 0.05 were considered statistically significant and are denoted by an asterisk (“*”).
Table 2. Disease etiology of the study population and subgroup sSema4D concentrations.
Table 2. Disease etiology of the study population and subgroup sSema4D concentrations.
Etiology of (Non-)Sepsis Critical Illness, n (%)Sepsis
n = 125
Non-Sepsis
n = 67
sSema4D (ng/mL)p
Pulmonary69 (55.2) 33.11 (9.99–240.00)0.059
Abdominal19 (15.2) 36.69 (11.12–160.27)
Urogenital10 (8) 34.51 (16.30–82.13)
Other27 (21.6) 44.62 (15.70–184.60)
Cardiocirculatory disorder 13 (19.4)40.38 (20.90–71.58)0.809
Respiratory failure 10 (14.9)34.22 (12.26–93.25)
Advanced liver disease 13 (19.4)41.74 (24.26–203.30)
Other 31 (46.3)39.12 (12.81–163.01)
The absolute numbers and percentages of the respective subgroup (in parentheses) or the median and range (in parentheses) are given. Significance of the differences between more than two groups was assessed using the Kruskal–Wallis test. p-values < 0.05 were considered statistically significant.
Table 3. Comorbidities and their influence on sSema4D levels.
Table 3. Comorbidities and their influence on sSema4D levels.
ComorbiditysSema4D Concentration in ng/mL, Median (Range)p
Diabetes (n = 50)36.53 (9.99–240.00)0.996
Liver cirrhosis (n = 20) 47.25 (15.14–163.01)0.020 *
Coronary artery disease (n = 63) 36.69 (12.26–160.27)0.882
Hypertension (n = 75) 35.38 (12.95–240.00)0.480
Chronic alcohol abuse (n = 25) 32.38 (15.14–163.01)0.144
Chronic obstructive lung disease (n = 25)32.79 (12.26–160.27)0.410
Active malignancy (n = 23) 31.93 (11.75–160.27)0.188
The median and range (in parentheses) are given unless indicated otherwise. Significance of the difference between groups was assessed using the Mann–Whitney U test. p-values < 0.05 were considered statistically significant and are denoted by an asterisk (“*”).
Table 4. Correlations of clinical and laboratory parameters with sSema4D serum concentrations at ICU admission.
Table 4. Correlations of clinical and laboratory parameters with sSema4D serum concentrations at ICU admission.
Parameterrp-Value
Demographics
Age−0.0950.191
BMI0.0610.406
Markers of inflammation and blood count
Leukocytes0.1440.048 *
Platelets0.0200.781
CRP0.0200.781
PCT0.0160.852
IL-6−0.0890.278
IL-100.1430.155
Markers of hepatobiliary function and coagulation
ALT0.0160.027 *
AST0.258<0.001 *
γGT0.2370.001 *
Bilirubin0.1700.019 *
Albumin−0.0130.888
Lipase−0.0090.917
INR0.0060.934
PTT0.0290.695
Electrolytes and renal function
Sodium−0.0300.677
Potassium0.1030.157
Urea0.0280.701
Creatinine0.0440.548
Cystatin C0.1200.185
Net fluid balance day 10.0010.986
Net fluid balance day 30.0080.921
Cardiopulmonary system
NT-pro-BNP−0.0060.953
Norephinephrine demand at day 1 (µg/day)−0.0840.257
Horovitz quotient (PaO2/FiO2)−0.0990.429
Ventilatory FiO2 demand0.1080.384
Metabolic markers
Glucose−0.0720.324
HbA1c−0.1210.260
Insulin−0.0080.940
C-Peptide0.0910.395
Total cholesterol0.0760.347
HDL−0.0080.943
LDL0.1310.228
Triglycerides0.2010.012 *
Clinical scores
Days on ICU−0.0720.322
SOFA score day 1−0.0400.727
SOFA score day 3−0.0330.809
APACHE-II day 10.0220.791
APACHE-II day 3−0.1570.224
A Spearman’s rank correlation test was used to calculate the significance of correlations of a positive and negative nature. p-values < 0.05 were considered statistically significant and are denoted by an asterisk (“*”). Abbreviations: ICU: intensive care unit; BMI: Body mass index; CRP: C-reactive protein; PCT: Procalcitonin; IL: Interleukin; ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; γGT: Gamma-glutamyl transpeptidase; INR: International normalized ratio; PTT: Partial Thromboplastin Time; NT-pro-BNP: N-terminal pro B-type natriuretic peptide; PaO2: Arterial oxygen partial pressure; FiO2: Fraction of inspired oxygen; HbA1c: Glycosylated hemoglobin A1; HDL: High-density lipoprotein; LDL: Low-density lipoprotein; SOFA: Sepsis-related Organ Failure Assessment; APACHE-II: Acute Physiology and Chronic Health Evaluation II.
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

Abu Jhaisha, S.; Hohlstein, P.; Yagmur, E.; Köller, V.; Pollmanns, M.R.; Adams, J.K.; Wirtz, T.H.; Brozat, J.F.; Bündgens, L.; Hamesch, K.; et al. Soluble Semaphorin 4D Serum Concentrations Are Elevated in Critically Ill Patients with Liver Cirrhosis and Correlate with Aminotransferases. Diagnostics 2024, 14, 370. https://doi.org/10.3390/diagnostics14040370

AMA Style

Abu Jhaisha S, Hohlstein P, Yagmur E, Köller V, Pollmanns MR, Adams JK, Wirtz TH, Brozat JF, Bündgens L, Hamesch K, et al. Soluble Semaphorin 4D Serum Concentrations Are Elevated in Critically Ill Patients with Liver Cirrhosis and Correlate with Aminotransferases. Diagnostics. 2024; 14(4):370. https://doi.org/10.3390/diagnostics14040370

Chicago/Turabian Style

Abu Jhaisha, Samira, Philipp Hohlstein, Eray Yagmur, Vera Köller, Maike R. Pollmanns, Jule K. Adams, Theresa H. Wirtz, Jonathan F. Brozat, Lukas Bündgens, Karim Hamesch, and et al. 2024. "Soluble Semaphorin 4D Serum Concentrations Are Elevated in Critically Ill Patients with Liver Cirrhosis and Correlate with Aminotransferases" Diagnostics 14, no. 4: 370. https://doi.org/10.3390/diagnostics14040370

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

Article Metrics

Back to TopTop