Skip to main content

sCD163, sCD28, sCD80, and sCTLA-4 as soluble marker candidates for detecting immunosenescence

Abstract

Background

Inflammaging, the characteristics of immunosenescence, characterized by continuous chronic inflammation that could not be resolved. It is not only affect older people but can also occur in young individuals, especially those suffering from chronic inflammatory conditions such as autoimmune disease, malignancy, or chronic infection. This condition led to altered immune function and as consequent immune function is reduced. Detection of immunosenescence has been done by examining the immune risk profile (IRP), which uses flow cytometry. These tests are not always available in health facilities, especially in developing countries and require fresh whole blood samples. Therefore, it is necessary to find biomarkers that can be tested using stored serum to make it easier to refer to the examination. Here we proposed an insight for soluble biomarkers which represented immune cells activities and exhaustion, namely sCD163, sCD28, sCD80, and sCTLA-4. Those markers were reported to be elevated in chronic diseases that caused early aging and easily detected from serum samples using ELISA method, unlike IRP. Therefore, we conclude these soluble markers are beneficial to predict pathological condition of immunosenescence.

Aim

To identify soluble biomarkers that could replace IRP for detecting immunosenescence.

Conclusion

Soluble costimulatory molecule suchsCD163, sCD28, sCD80, and sCTLA-4 are potential biomarkers for detecting immunosenescence.

Background

The concept of “inflammaging” introduced by Franceschi et al., (2000), was related to declining ability of immune system to cope with various external stressors, as well as accelerating pro-inflammatory status. They proposed that continuous exposure to internal and external stressors led to chronic macrophage activation, naïve immune cells exhaustion, decreased in the T-cell repertoire and affected other immune cells. These continuous disruptions over the time resulting a major change and depleted the ability of immune system to response against stressors, as well as escalating pro-inflammatory response. These conditions define the characteristic of immunosenescence [1].

Immunosenescence was commonly identified with immune risk profile (IRP). The concept of IRP has originated from the OCTO/NONA study conducted in Sweden in individuals over 85 years of age. IRP is an immunosenescence marker characterized by the presence of 100% cytomegalovirus (CMV) infection and a CD4/CD8 ratio < 1 owing to the accumulation of differentiated T cells, particularly CD27CD28CD57CD8+ T cells. These are the antigen-specific T cells for CMV [2,3,4]. Furthermore, IRP is characterized by a lower number of CD8+CD45RA+ cells and higher number of CD8+CD45RO+ T cells [3, 5, 6].

An inverted CD4/CD8 ratio (< 1) is associated with an increase in the number of activated, senescent, and exhausted CD4+ T cells and CD8+ T cells, as well as a shift in naïve to memory cells [7]. In addition, an inverted CD4/CD8 ratio indicates accumulation of CD8+ T cells that have differentiated into the late phase, a low proliferative response of T cells, and a low number of B cells [5]. An inverted CD4/CD8 ratio has been linked with premature immunosenescence in individuals of all ages, including children and young adults infected with Human Immunodeficiency Virus (HIV), patients with myocardial infarction, and patients under physical and psychological stress. The prevalence of an inverted CD4/CD8 ratio is 8% between the age of 20 and 59 and up to 16% between the age of 60 and 94. The inverted CD4/CD8 ratio correlates with an increased mortality and persistent viral infection [8].

In addition to the inverted CD4/CD8 ratio, one of the IRPs is a decrease in T-cells with CD28 receptors. CD28 is a costimulatory molecule responsible for activating T cells. However, during activation, some T cells lose the CD28 molecule and become CD28 T cells. These CD28 T cells act as antigen-recognizing cells and are highly differentiated. During normal aging, CD8+CD28 T cells are accumulated, which is likely due to the continuous exposure to various antigens present through lifetime [9]. A study by Yadav et al. found that CD4+CD28 T cells in patients with chronic kidney disease (CKD) were correlated with an increased occurrence of atherosclerosis [10]. CD4+CD28 T cells also show increased cytotoxic and inflammatory activity. Téo et al. showed that CD4+CD28 T cells participate in the pathogenesis of atherosclerosis and their number also increases in patients with acute coronary syndrome (ACS) [11].

An increase in memory T cells (CD4+CD45RO+ T cells and CD8+CD45RO+ T cells) and a decrease in naïve T cells (CD4+CD45RA+ T cells and CD8+CD45RA+ T cells) is observed during immunosenescence. Memory T cells increase with age and are more abundant in tissues [12]. Naïve T cells (CD45RA+) express CD27, CD28, and CCR7 as they leave the thymus. When exposed to antigens, naïve T cells differentiate into central memory T cells (CD45RO+ T cells) [13]. Involution of the thymus leads to a decrease in naïve T cells. These conditions make geriatric population more susceptible to new antigens [14].

In addition to these parameters, IRP is also characterized by the presence of CMV infection. CMV mainly survives in myeloid cells but can sometimes be found in other cells. When CMV infection occurs, containment of the wider infection is a priority for the immune system; however, complete elimination is never achieved. A study found changes in CD8+ T cells that were very similar to senescence but occurred as a result of CMV infection [15]. The CMV seropositive parameter can predict mortality in the geriatric population. These findings indicate the presence of persistent CMV as a chronic antigen stressor, which is a major contributor to immunosenescence and mortality [15, 16].

Although many IRPs have been identified, which can be used for the detection of immunosenescence, these markers are cell surface receptors that have to be examined using flow cytometry, which required a fresh blood sample. The required condition is difficult to accommodate especially in developing countries, where most of the healthcare facilities lack of advance instruments, such as flow cytometry. As a result, immunosenescence markers in the dissolved form are required. Markers in the dissolved form are more stable and could be measured from stored serum. Other promising soluble marker candidates for accelerated aging, such as soluble urokinase plasminogen activator receptor (suPAR) was not easily detected, as it was present in low concentration in the serum [17]. Other soluble markers such as IL-6, TNF-α and IFN-γ were also commonly used as biomarkers for immunosenescence detection, however those markers were less specific as it can be also detected during acute inflammation [18,19,20].

This review focused on four different soluble markers, CD163, CD28, CD80 and CTLA-4, which were commonly known to have various functions related to macrophage [21,22,23] and T cells activation [24,25,26], which play a key role in immunosenescence. Characterization of CD163 and CD80 biomarkers, which determine the macrophage polarization, had been used as to monitor inflammaging [27].

While CD28 is linked to inflammaging as it is observed to be absent in elder people as well as age-related diseases. The senescence is associated with an increase of CD28- memory T cell and decrease of naïve T cell populations, therefore the CD28 could be a good biomarker candidate of immunosenescence [28]. CD28- T cells are also known to have a short telomerase, as a consequence it will affect negatively on the immune checkpoint inhibitory receptors, such as CTLA-4. Suppressive function of Treg was regulated by CTLA-4 receptor together with the co-stimulatory receptor CD28. Therefore, CTLA-4, together with PD-1, which regulate the T cells response, play important roles in maintaining the balance between stimulatory and inhibitory signals for immune responses against antigens [28, 29], thus determine the senescence status.

Moreover, the soluble markers mentioned above were easily detected using enzyme-linked immunosorbent assay (ELISA) method, as they were present in high concentration in the serum [30,31,32,33].

Soluble CD163

CD163 is a glycosylated membrane protein [23] expressed almost exclusively on all macrophages [21,22,23] and at least 10-30% of monocytes [34]. CD163 is a member of the B scavenger receptor cysteine-rich (SRCR) family [22] and consists of 9 extracellular SRCR protein domains associated with the short transmembrane segment and short cytoplasmic tail. CD163 is located on chromosome 12p13 and consists of 17 exons [23]. CD163 is also a scavenger hemoglobin (Hb) receptor involved in the endocytosis of the haptoglobin-Hb complex [21, 23]. It has other functions as well, including those related to erythroblast adhesion, immune sensing of the presence of bacteria, and bonding with TNF-like weak inducer of apoptosis (TWEAK) [23]. In vitro, CD163 expression is increased by the presence of glucocorticoids, IL-10 and IL-6 but not by IL-4 or IL-13, but not by IL-4 and IL-13 [21, 23]. CD163 expression is reduced by the presence of tumors necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and chemokine CXCL4 (platelet factor 4). This suggests that CD163 is mainly expressed by M2 macrophages [23].

CD163 can be released (shedding) upon the action of cleaving enzyme TNF-α [35], resulting in a dissolved form known as soluble CD163 (sCD163) [23, 35]. The sCD163 level is negatively correlated with the level of membrane bound CD163 [35]. sCD163 has been detected in the plasma of healthy individuals at levels ranging from 0.73 to 4.69 mg/L, with a median of 1.87 mg/L [36]. Because its expression is limited to the monocyte line, sCD163 can be used as a specific marker of macrophage and monocyte activation [22, 35]. Furthermore, sCD163 and non-membrane-bound products function similarly to cytokines in that they inhibit the activation and proliferation of T lymphocytes [35], particularly CD4+ T cells [36]. The release of sCD163 by cleaving enzyme TNF-α corresponds to an increase in sCD163 levels in acute and chronic inflammatory diseases [35, 37] and hematological diseases [35].

In vivo, the release of sCD163 occurred simultaneously with an increase in TNF-α response due to the presence of lipopolysaccharide (LPS) mediated by Toll-like Receptors (TLR) 4 activation. Then, it is accompanied by a sharp increase in sCD163 and TNF-α levels and positively correlated with the number and activity of monocytes or macrophages (Fig. 1) [23, 38]. In addition, sCD163 was released in the presence of oxidants and pro-inflammatory cytokines [39]. The half-life of sCD163 is much longer than TNF-α and sCD163 levels remain elevated for 1–2 days [23, 40]. Levels of sCD163 can be affected by several factors such as increased CD163 expression, increased shedding, and impaired clearance [23]. In healthy individuals, sCD163 levels were shown to have low individual variability with a value limit of ± 30% [35].

Fig. 1
figure 1

Shedding of soluble CD163. Various inflammatory signals induce shedding of sCD163 in vitro. Shedding of sCD163 can be induced by TLR4 activation or FcγR-crosslinking mediated by ADAM17/TACE. Half-life of sCD163 is longer than that of TNF-α. FcγR, Fc-gamma receptors; LPS, Lipopolysaccharides; TACE/ADAM17, Tumor necrosis factor α-converting enzyme; TLR, Toll-like receptors; TNF, Tumor necrosis factor. Reprinted with permission from Reference: Møller, H. J. 2012. Soluble CD163. Scandinavian journal of clinical and laboratory investigation, 72, 1–13

Various studies have been conducted to better understand sCD163; however, research on sCD163 in the context of immunosenescence in geriatrics is lacking. Previous studies have shown that sCD163 is elevated in patients with HIV and is linked with the aging of CD8+ T cells in these patients. Researchers have discovered that sCD163 levels in patients with HIV were equivalent to those of 14.5 year older healthy individuals. Furthermore, an increase in sCD163 with age has been shown [41]. Other HIV-related studies have demonstrated a negative relationship between sCD163 levels and telomere length. During HIV infection, telomere shortening occurs, which is a process associated with aging. Increased sCD163 in patients with HIV infection also correlates with the emergence of co-morbidities in the form of premature degenerative diseases such as cardiovascular and neurological diseases [42, 43]. This indicates a correlation between chronic immune system activation and increased sCD163 levels.

Hodowanec et al. found that sCD163 levels positively correlated with anti-CMV IgG antibody levels were positively correlated in patients with HIV infection [44]. Azanan et al. found an increase in sCD163 in pediatric patients with leukemia when compared with healthy children of the same age; sCD163 levels correlated with anti-CMV IgG antibodies in these patients [45]. Another study found an increase in sCD163 in patients with chronic Hepatitis C Virus (HCV) infection. sCD163 is also associated with increased mortality from heart disease, acute myocardial infarction, increased mortality, atherosclerosis, diabetes, and insulin resistance [46].

Various studies regarding sCD163 levels in serum and urine samples of various chronic diseases are listed in Table 1. In addition, Zhi et al. analyzed the role of sCD163 in asthma. According to their study, sCD163 plays a role in the pathogenesis of asthma and can act as a potential marker as well as a target for therapy [47]. Meanwhile, the meta-analysis by Qian et al. suggested that sCD163 is correlated with the risk of mortality in cancer [48].

Based on Table 1, it can be concluded that sCD163 levels, especially serum sCD163, correlate with various chronic inflammatory conditions arising due to infection, autoimmunity, or malignancy. Chronic inflammatory conditions are similar to inflammaging in immunosenescence. However, to the best of our knowledge, no studies have reported the role of sCD163 in immunosenescence.

Table 1 The role of sCD163 in various chronic diseases

Soluble CD28

CD28 is a T cell surface receptor that strengthens the transcriptional effect of TCR and acts as a costimulatory receptor for naïve T cell activation [70]. CD28 is expressed in 95% CD4+ T cells and 50% human CD8+ T cells but number of CD28+ T cells decrease during aging and in the presence of CMV infection [71, 72]. CD28 can bind to receptors on antigen-presenting cells (APC), CD80 (B7-1), and CD86 (B7-2) for T cell activation (Fig. 2A and B). Binding of CD28 with its ligand also triggers anti-apoptosis, increases cytokine secretion, especially that of IL-2, increases cell adhesion, prevents the induction of T cell anergy, and triggers the formation of a germinal center [73]. CD28 deficiency leads to impaired T cell proliferation, changes in immunoglobulin class, germinal center formation, and impaired Th2 cell response [74, 75]. CD28 is abundant in naïve T cells but in highly differentiated T cells, number of CD28 significantly decreased [9, 13]. T cell activation occurs through its binding with membrane-bound CD28, which is followed by the shedding of membrane-bound CD28 in the plasma, referred to as soluble CD28 (sCD28). CD28 functions in T cell regulation while being expressed on the cell surface as well as in its dissolved form [76].

Soluble CD28 originates from released surface membrane receptors or alternative mRNA splicing (Fig. 2B) [31, 77]. However, recent studies using RT-PCR analysis have shown that in systemic lupus erythematosus (SLE) patients, an increase in sCD28 levels results from shedding of membrane-bound CD28 [78]. This is consistent with a study by Sun et al., who assessed sCD28 levels in Graves’ disease and found that an increase in sCD28 levels correlated with a decrease in membrane-bound CD28 [31]. In vitro, sCD28 stimulates T cell proliferation as well as IL-6 and TNF-secretion. In vivo, sCD28 serves as a marker for increased CD28 expression on T cells, indicating APC and T cell activation. Furthermore, sCD28 can compete and interfere with the interaction of CD28 or CTLA-4 with B7 (Fig. 2F) [31]. sCD28 has been found to be elevated in a variety of autoimmune diseases, including SLE and rheumatoid arthritis (RA) [76, 79] as detailed in Table 2. In addition, elevated sCD28 levels are also associated with chronic inflammatory conditions such as malignancy, chronic infection, and metabolic disorders (diabetes mellitus) (Table 2).

Table 2 Role of sCD28 in various chronic disease

Based on Table 2, sCD28 plays a role in a variety of diseases, particularly autoimmune disorders, malignancy, and chronic infections. There is a common pathogenesis in these three types of diseases in terms of chronic low-grade inflammation, which corresponds to an inflammaging condition. As a result, the authors conclude that sCD28 plays a role in inflammaging, including immunosenescence. A strong negative correlation also exists between sCD28 levels and number of CD28+ T cells. This demonstrates that immune system activation causes CD28 to be shed into its soluble form. Therefore, in immunosenescence (which is associated with inflammaging), an increase in sCD28 levels is likely to be observed.

Soluble CD80

CD80 is a costimulatory factor expressed on the surface of activated monocytes, B cells, and dendritic cells [99]. CD80 binds to CD28 to activate T cells (Fig. 2B). CD80 expression is stimulated by APC; however, small amounts of CD80 are expressed on inactive monocytes. The soluble form of CD80, soluble CD80 (sCD80), originates from spliced mRNA or the release of cell surface CD80 receptor into the circulation [25]. The spliced form of sCD80 is expressed by inactivated monocytes and B cells [99]. CD80 prevents programmed death-ligand-1 (PD-L1) mediated immune suppression and PD-1 in tumor cells. sCD80 has the same strong ability to bind PD-L1 as that of CD80, such that sCD80 suppresses PD-L1 function. In addition, sCD80 is also able to bind CD28 and cytotoxic T lymphocyte-associated molecule-4 (CTLA-4) (Fig. 2C and E) [24, 99, 100]. When it binds to CD28, sCD80 activates T cells (Fig. 2C). However, when it binds to CTLA-4, sCD80 does not lead to either T cell suppression or activation, indicating that CTLA-4 is a receptor that functions as a decoy and does not have a biological function of T cell suppression (Fig. 2E) [24]. A study found that an increase in sCD80 levels leads to an increase in IFN-γ production by active T cells [99].

sCD80 levels were found to be > 15 µg/L in 24% of healthy individuals. However, sCD80 levels increased significantly in patients with SLE and leukemia compared to the healthy population [25]. A study found that sCD80 could prevent PD-L1 suppression and restore T cell activation by blocking interaction with PD-L1. In mice, sCD80 can slow tumor growth and trigger T cells to infiltrate tumor cells in vivo. The study concluded that sCD80 can act as a therapeutic agent to slow tumor growth [24, 100, 101]. Apart from malignancy, sCD80 also plays a role in other chronic diseases, such as minimal change disease (MCD) in adult humans, as its level in urine samples were known to increase, but not in serum. It is thought that sCD80 plays a role in the pathogenesis of MCD [102]. In addition, various studies have also found increased levels of sCD80 in numerous diseases as shown in Table 3.

Table 3 The role of sCD80 level in various chronic disease

Based on Table 3, it can be concluded that sCD80 level in the blood is increased in autoimmune diseases, allergies, chronic infections, and malignancies. In kidney disease, an increase in blood sCD80 level is not observed, but the levels are increased in the urine. sCD80 has so far been known as an immune checkpoint against malignancy; however, further research indicates its roles beyond that in malignancy. The authors envisage that in chronic inflammatory conditions, the elevation in sCD80 levels is caused by excess T cell activation, known as inflammaging. However, no study has reported the association of sCD80 levels with inflammaging that occurs during immunosenescence.

Soluble CTLA-4

Cytotoxic T lymphocyte-associated molecule-4 or CD152 is a receptor found on T cells and plays an important role in the regulation of the immune system. CTLA-4 is homologous to CD28 and can bind to the same ligand, namely CD80/CD86 on APC (Fig. 2A) [116, 117]. CTLA-4 competes with CD28 for binding with CD80 and CD86. However, contrary to popular belief, binding of CTLA-4 to CD80/CD86 has no suppressive effect on T cells; instead, CTLA-4 acts as a decoy receptor to prevent T cell activation (Fig. 2D) [24]. CTLA-4 is strongly stimulated by activated T and B cells, and is also expressed on striated muscle cells and placental fibroblasts. In addition, 3% monocyte also express CTLA-4 on their cell surface and 20% monocytes express intracellular CTLA-4. In vitro, when stimulated by IFN-γ, monocytes secrete the soluble form of CTLA-4, soluble CTLA-4 (sCTLA-4) [117]. In vivo, sCTLA-4 is formed from alternatively spliced mRNA or comes from shedding membrane-bound CTLA-4 (Fig. 2D). Furthermore, sCTLA-4 can also be produced by T cells, especially Treg, in vitro. sCTLA-4 transcripts have been detected in lymph nodes, spleen, CD4 and CD8 T cells, B cells, and monocytes [116, 118, 119].

Several studies have found an increase in sCTLA-4 levels in autoimmune diseases such as Graves’ disease, Hashimoto’s thyroiditis, myasthenia gravis, SLE, type I DM, celiac disease, systemic sclerosis, and autoimmune pancreatitis disease. As a result, it can be concluded that sCTLA-4 levels play an important regulatory role in the immune system. sCTLA-4, on the other hand, can interfere with the interaction of CD80 or CD86 with CTLA-4, thereby blocking negative CTLA-4 signals (Fig. 2F) [116, 118,119,120]. Another in vitro study on melanoma cancer cells found that these cells could produce CTLA-4 and sCTLA-4, indicating a possible role of CTLA-4 and sCTLA-4 in cancer growth [33]. In addition, other studies have found that anti-CTLA-4 antibodies can bind sCTLA-4; further, sCTLA-4 has been shown to induce an antitumor response and has been suggested as an alternative therapeutic option for melanoma [99, 119]. A study in geriatric population found a positive correlation between levels of sCTLA-4 and pro-inflammatory cytokines [121]. Many studies have been conducted regarding the role of sCTLA-4 in various diseases as summarized in Table 4.

Fig. 2
figure 2

Interaction between APC receptor (CD80 and CD86) with T cell receptor (CD28 and CTLA-4). (A) Both CD80 and CD86 can bind to CD28 or CTLA-4. (B) Binding of CD80 or CD86 to CD28 will activate T cells also shedding of CD28 (become sCD28). (C) Binding of sCD80 to CD28 will activate T cells. (D) Binding of CD80 or CD86 to CTLA-4 will suppress T cells also shedding of CTLA-4 (become sCTLA-4). (E) Binding of sCD80 to CTLA-4 will suppress T cells. (F) If CD80 or CD86 bind to either sCD28 or sCTLA-4, T cells will have no response and become anergy

Table 4 The role of sCTLA-4 level in various chronic disease

According to Table 4, sCTLA-4 plays a role in autoimmune disorders cancer, chronic infection, and a variety of other chronic inflammatory conditions. Because chronic inflammation is a feature of immunosenescence, the authors hypothesize that there is an increase in sCTLA-4 levels during immunosenescence. However, no research on sCTLA-4 in the context of immunosenescence in the elderly has ever been conducted.

Conclusion

Soluble markers sCD163, sCD28, sCD80, and sCTLA-4 have promising potential to confirm the pathological condition of immunosenescence. These soluble markers were detectable in high level in the serum samples using ELISA method, thus might be and might be used to replace the cell surface receptors as immunosenescence markers. However, further research comparing their diagnostic performance with the gold standard (cell surface receptor) assay is required to make a highly accurate prediction model of immunosenensence.

References

  1. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-aging: an evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908(1):244–54.

    Article  CAS  PubMed  Google Scholar 

  2. Pawelec G, Larbi A, Derhovanessian E. Senescence of the human immune system. J Comp Pathol. 2010;142:39–S44.

    Article  Google Scholar 

  3. Wikby A, Strindhall J, Johansson B. The immune risk profile and associated parameters in late life: lessons from the OCTO and NONA longitudinal studies. Handbook on Immunosenescence: Springer; 2009. pp. 3–28.

    Google Scholar 

  4. Chen X, Liu Q, Xiang AP. CD8 + CD28-T cells: not only age-related cells but a subset of regulatory T cells. Cell Mol Immunol. 2018;15(8):734–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Derhovanessian E, Maier AB, Beck R, Jahn G, Hähnel K, Slagboom PE, et al. Hallmark features of immunosenescence are absent in familial longevity. J Immunol. 2010;185(8):4618–24.

    Article  CAS  PubMed  Google Scholar 

  6. Xydonas S, Parissis J, Lioni L, Kapsimali V, Psarra E, Farmakis D, et al. Immunosenescence in patients with chronic systolic Heart Failure. J Cardiovasc Med. 2016;17(8):624–30.

    Article  CAS  Google Scholar 

  7. Sainz T, Serrano-Villar S, Díaz L, Tomé MIG, Gurbindo MD, de José MI, et al. The CD4/CD8 ratio as a marker T-cell activation, senescence and activation/exhaustion in treated HIV-infected children and young adults. Aids. 2013;27(9):1513–6.

    Article  CAS  PubMed  Google Scholar 

  8. Muller GC, Gottlieb MGV, Correa BL, Gomes Filho I, Moresco RN, Bauer ME. The inverted CD4: CD8 ratio is associated with gender-related changes in oxidative stress during aging. Cell Immunol. 2015;296(2):149–54.

    Article  CAS  PubMed  Google Scholar 

  9. Mou D, Espinosa J, Lo DJ, Kirk AD. CD28 negative T cells: is their loss our gain? Am J Transplant. 2014;14(11):2460–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yadav AK, Banerjee D, Lal A, Jha V. Vitamin D deficiency, CD4 + CD28null cells and accelerated Atherosclerosis in chronic Kidney Disease. Nephrology. 2012;17(6):575–81.

    Article  CAS  PubMed  Google Scholar 

  11. Téo FH, de Oliveira RTD, Mamoni RL, Ferreira MCS, Nadruz W Jr, Coelho OR, et al. Characterization of CD4 + CD28null T cells in patients with coronary artery Disease and individuals with risk factors for Atherosclerosis. Cell Immunol. 2013;281(1):11–9.

    Article  PubMed  Google Scholar 

  12. Montoya-Ortiz G. Immunosenescence, aging, and systemic lupus erythematous. Autoimmune diseases. 2013;2013.

  13. van den Laurens L, Patrick Sims G, van Adrianus Gijsbert J. Dorothea Elisabeth Fritsch-Stork R. Aging and systemic lupus erythematosus-immunosenescence and beyond. Curr Aging Sci. 2015;8(2):158–77.

    Article  Google Scholar 

  14. Lang PO, Govind S, Aspinall R. Reversing T cell immunosenescence: why, who, and how. Age. 2013;35(3):609–20.

    Article  PubMed  Google Scholar 

  15. Koch S, Solana R, Rosa OD, Pawelec G. Human cytomegalovirus Infection and T cell immunosenescence: a mini review. Mech Ageing Dev. 2006;127(6):538–43.

    Article  CAS  PubMed  Google Scholar 

  16. Pawelec G, Derhovanessian E, Larbi A, Strindhall J, Wikby A. Cytomegalovirus and human immunosenescence. Rev Med Virol. 2009;19(1):47–56.

    Article  CAS  PubMed  Google Scholar 

  17. Rasmussen LJH, Caspi A, Ambler A, Danese A, Elliott M, Eugen-Olsen J, et al. Association between elevated suPAR, a new biomarker of inflammation, and accelerated aging. The Journals of Gerontology: Series A. 2021;76(2):318–27.

    CAS  Google Scholar 

  18. Del Giudice M, Gangestad SW, Rethinking. IL-6 and CRP: why they are more than inflammatory biomarkers, and why it matters. Brain Behav Immun. 2018;70:61–75.

    Article  PubMed  Google Scholar 

  19. Zelová H, Hošek J. TNF-α signalling and inflammation: interactions between old acquaintances. Inflamm Res. 2013;62:641–51.

    Article  PubMed  Google Scholar 

  20. Kak G, Raza M, Tiwari BK. Interferon-gamma (IFN-γ): exploring its implications in infectious Diseases. Biomol Concepts. 2018;9(1):64–79.

    Article  CAS  PubMed  Google Scholar 

  21. Gaini S, Pedersen S, Koldkjær O, Pedersen C, Moestrup S, Møller H. New immunological serum markers in bacteraemia: anti-inflammatory soluble CD163, but not proinflammatory high mobility group-box 1 protein, is related to prognosis. Clin Experimental Immunol. 2008;151(3):423–31.

    Article  CAS  Google Scholar 

  22. Møller H, Aerts H, Grøbæk H, Peterslund N, Petersen PH, Hornung N, et al. Soluble CD163: a marker molecule for monocyte/macrophage activity in Disease. Scand J Clin Lab Investig. 2002;62(7):29–33.

    Article  Google Scholar 

  23. Møller HJ. Soluble CD163. Scand J Clin Lab Investig. 2012;72(1):1–13.

    Article  Google Scholar 

  24. Horn LA, Long TM, Atkinson R, Clements V, Ostrand-Rosenberg S. Soluble CD80 protein delays Tumor growth and promotes tumor-infiltrating lymphocytes. Cancer Immunol Res. 2018;6(1):59–68.

    Article  CAS  PubMed  Google Scholar 

  25. Kakoulidou M, Giscombe R, Zhao X, Lefvert A, Wang X. Human soluble CD80 is generated by alternative splicing, and recombinant soluble CD80 binds to CD28 and CD152 influencing T-cell activation. Scand J Immunol. 2007;66(5):529–37.

    Article  CAS  PubMed  Google Scholar 

  26. Xia F, Qian C-R, Xun Z, Hamon Y, Sartre A-M, Formisano A, et al. TCR and CD28 concomitant stimulation elicits a distinctive calcium response in naive T cells. Front Immunol. 2018;9:2864.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Costantini A, Viola N, Berretta A, Galeazzi R, Matacchione G, Sabbatinelli J, et al. Age-related M1/M2 phenotype changes in circulating monocytes from healthy/unhealthy individuals. Aging. 2018;10(6):1268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Aiello A, Farzaneh F, Candore G, Caruso C, Davinelli S, Gambino CM, et al. Immunosenescence and its hallmarks: how to oppose aging strategically? A review of potential options for therapeutic intervention. Front Immunol. 2019;10:2247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Santoro A, Bientinesi E, Monti D. Immunosenescence and inflammaging in the aging process: age-related Diseases or longevity? Ageing Res Rev. 2021;71:101422.

    Article  CAS  PubMed  Google Scholar 

  30. Nielsen MC, Andersen MN, Rittig N, Rødgaard-Hansen S, Grønbæk H, Moestrup SK, et al. The macrophage-related biomarkers sCD163 and sCD206 are released by different shedding mechanisms. J Leukoc Biol. 2019;106(5):1129–38.

    Article  CAS  PubMed  Google Scholar 

  31. Sun Z, Yi L, Tao H, Huang J, Jin Z, Xiao Y, et al. Enhancement of soluble CD28 levels in the serum of Graves’ Disease. Central-European J Immunol. 2014;39(2):216.

    Article  Google Scholar 

  32. Matsuyama Y, Asanuma K, Yoshida K, Hagi T, Iino T, Nakamura T, et al. The role of soluble CD80 in patients with soft tissue tumors. J Orthop Surg Res. 2022;17(1):1–9.

    Article  Google Scholar 

  33. Khanolkar RC, Zhang C, Al-Fatyan F, Lawson L, Depasquale I, Meredith FM, et al. TGFβ2 induces the Soluble Isoform of CTLA-4–Implications for CTLA-4 based checkpoint inhibitor antibodies in malignant Melanoma. Front Immunol. 2022;12:5687.

    Article  Google Scholar 

  34. Matsushita N, Kashiwagi M, Wait R, Nagayoshi R, Nakamura M, Matsuda T, et al. Elevated levels of soluble CD163 in sera and fluids from rheumatoid arthritis patients and inhibition of the shedding of CD163 by TIMP-3. Clin Experimental Immunol. 2002;130(1):156–61.

    Article  CAS  Google Scholar 

  35. Davis BH, Zarev PV. Human monocyte CD163 expression inversely correlates with soluble CD163 plasma levels. Cytometry Part B: Clinical Cytometry: The Journal of the International Society for Analytical Cytology. 2005;63(1):16–22.

    Article  Google Scholar 

  36. Onofre G, Kolácková M, Jankovicová K, Krejsek J. Scavenger receptor CD163 and its biological functions. Acta Medica (Hradec Kralove). 2009;52(2):57–61.

    Article  CAS  PubMed  Google Scholar 

  37. Etzerodt A, Moestrup SK. CD163 and inflammation: biological, diagnostic, and therapeutic aspects. Antioxid Redox Signal. 2013;18(17):2352–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Weaver LK, Hintz-Goldstein KA, Pioli PA, Wardwell K, Qureshi N, Vogel SN, et al. Pivotal advance: activation of cell surface toll‐like receptors causes shedding of the hemoglobin scavenger receptor CD163. J Leukoc Biol. 2006;80(1):26–35.

    Article  CAS  PubMed  Google Scholar 

  39. Moreno JA, Ortega-Gomez A, Delbosc S, Beaufort N, Sorbets E, Louedec L, et al. In vitro and in vivo evidence for the role of elastase shedding of CD163 in human atherothrombosis. Eur Heart J. 2012;33(2):252–63.

    Article  CAS  PubMed  Google Scholar 

  40. Nielsen MC, Andersen MN, Rittig N, Rødgaard-Hansen S, Grønbæk H, Moestrup SK, et al. The macrophage‐related biomarkers sCD163 and sCD206 are released by different shedding mechanisms. J Leukoc Biol. 2019;106(5):1129–38.

    Article  CAS  PubMed  Google Scholar 

  41. Martin GE, Gouillou M, Hearps AC, Angelovich TA, Cheng AC, Lynch F, et al. Age-associated changes in monocyte and innate immune activation markers occur more rapidly in HIV infected women. PLoS ONE. 2013;8(1):e55279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Srinivasa S, Fitch KV, Petrow E, Burdo TH, Williams KC, Lo J, et al. Soluble CD163 is associated with shortened telomere length in HIV-infected patients. J Acquir Immune Defic Syndr. 2014;67(4):414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Burdo TH, Weiffenbach A, Woods SP, Letendre S, Ellis RJ, Williams KC. Elevated sCD163 in plasma but not cerebrospinal fluid is a marker of neurocognitive impairment in HIV Infection. AIDS. 2013;27(9).

  44. Hodowanec A, Williams B, Hanson B, Livak B, Keating S, Lurain N, et al. Soluble CD163 but not soluble CD14 is associated with cytomegalovirus immunoglobulin G antibody levels in virologically suppressed HIV + individuals. JAIDS J Acquir Immune Defic Syndr. 2015;70(5):e171–e4.

    Article  PubMed  Google Scholar 

  45. Azanan MS, Abdullah NK, Chua LL, Lum SH, Abdul Ghafar SS, Kamarulzaman A, et al. Immunity in young adult survivors of childhood Leukemia is similar to the elderly rather than age-matched controls: role of cytomegalovirus. Eur J Immunol. 2016;46(7):1715–26.

    Article  CAS  PubMed  Google Scholar 

  46. Naggie S, Hepatitis C. Virus, inflammation, and cellular aging: turning back time. Top Antiviral Med. 2017;25(1):3.

    Google Scholar 

  47. Zhi Y, Gao P, Xin X, Li W, Ji L, Zhang L, et al. Clinical significance of sCD163 and its possible role in Asthma. Mol Med Rep. 2017;15(5):2931–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Qian S, Zhang H, Dai H, Ma B, Tian F, Jiang P, et al. Is sCD163 a clinical significant prognostic value in cancers? A systematic review and meta-analysis. Front Oncol. 2020;10:585297.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Aristoteli LP, Møller HJ, Bailey B, Moestrup SK, Kritharides L. The monocytic lineage specific soluble CD163 is a plasma marker of coronary Atherosclerosis. Atherosclerosis. 2006;184(2):342–7.

    Article  CAS  PubMed  Google Scholar 

  50. Hiraoka A, Horiike N, Akbar S, Fazle M, Michitaka K, Matsuyama T, et al. Soluble CD163 in patients with Liver Diseases: very high levels of soluble CD163 in patients with fulminant hepatic failure. J Gastroenterol. 2005;40(1):52–6.

    Article  CAS  PubMed  Google Scholar 

  51. Sandahl T, McGrail R, Møller H, Reverter E, Møller S, Turon F, et al. The macrophage activation marker sCD 163 combined with markers of the enhanced liver fibrosis (ELF) score predicts clinically significant portal Hypertension in patients with Cirrhosis. Aliment Pharmacol Ther. 2016;43(11):1222–31.

    Article  CAS  PubMed  Google Scholar 

  52. Andersen E, Rødgaard-Hansen S, Moessner B, Christensen PB, Møller H, Weis N. Macrophage-related serum biomarkers soluble CD163 (sCD163) and soluble mannose receptor (sMR) to differentiate mild liver fibrosis from Cirrhosis in patients with chronic Hepatitis C: a pilot study. Eur J Clin Microbiol Infect Dis. 2014;33(1):117–22.

    Article  CAS  PubMed  Google Scholar 

  53. Kazankov K, Barrera F, Møller HJ, Rosso C, Bugianesi E, David E, et al. The macrophage activation marker sCD 163 is associated with morphological Disease stages in patients with non-alcoholic fatty Liver Disease. Liver Int. 2016;36(10):1549–57.

    Article  CAS  PubMed  Google Scholar 

  54. Parkner T, Sørensen L, Nielsen A, Fischer C, Bibby B, Nielsen S, et al. Soluble CD163: a biomarker linking macrophages and insulin resistance. Diabetologia. 2012;55(6):1856–62.

    Article  CAS  PubMed  Google Scholar 

  55. Axelsson J, Møller HJ, Witasp A, Qureshi AR, Carrero JJ, Heimbürger O, et al. Changes in fat mass correlate with changes in soluble sCD163, a marker of mature macrophages, in patients with CKD. Am J Kidney Dis. 2006;48(6):916–25.

    Article  CAS  PubMed  Google Scholar 

  56. Castley A, Williams L, James I, Guelfi G, Berry C, Nolan D. Plasma CXCL10, sCD163 and sCD14 levels have distinct associations with antiretroviral treatment and Cardiovascular Disease risk factors. PLoS ONE. 2016;11(6):e0158169.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Beltran LM, Muñoz Hernández R, de Pablo Bernal RS, Garcia Morillo JS, Egido J, Noval ML, et al. Reduced sTWEAK and increased sCD163 levels in HIV-infected patients: modulation by antiretroviral treatment, HIV replication and HCV co-infection. PLoS ONE. 2014;9(3):e90541.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Silva RLL, Santos MB, Almeida PL, Barros TS, Magalhães L, Cazzaniga RA, et al. sCD163 levels as a biomarker of Disease severity in Leprosy and visceral Leishmaniasis. PLoS Negl Trop Dis. 2017;11(3):e0005486.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Grønbaek H, Kreutzfeldt M, Kazankov K, Jessen N, Sandahl T, Hamilton-Dutoit S, et al. Single‐centre experience of the macrophage activation marker soluble (s) CD163–associations with Disease activity and treatment response in patients with autoimmune hepatitis. Aliment Pharmacol Ther. 2016;44(10):1062–70.

    Article  PubMed  Google Scholar 

  60. Nishino A, Katsumata Y, Kawasumi H, Hirahara S, Kawaguchi Y, Yamanaka H. Usefulness of soluble CD163 as a biomarker for macrophage activation syndrome associated with systemic Lupus Erythematosus. Lupus. 2019;28(8):986–94.

    Article  CAS  PubMed  Google Scholar 

  61. David C, Divard G, Abbas R, Escoubet B, Chezel J, Chauveheid M, et al. Soluble CD163 is a biomarker for accelerated Atherosclerosis in systemic Lupus Erythematosus patients at apparent low risk for Cardiovascular Disease. Scand J Rheumatol. 2020;49(1):33–7.

    Article  CAS  PubMed  Google Scholar 

  62. Yang G, Guo N, Yin J, Wu J. Elevated soluble CD163 predicts renal function deterioration in lupus Nephritis: a cohort study in eastern China. J Int Med Res. 2021;49(11):03000605211049963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gupta R, Yadav A, Aggarwal A. Urinary soluble CD163 is a good biomarker for renal Disease activity in lupus Nephritis. Clin Rheumatol. 2021;40(3):941–8.

    Article  PubMed  Google Scholar 

  64. Gamal NM, Badawy ER, Talaat EA, Ibrahim HM, Abd Elsamea MH. Clinical utility of urinary soluble CD163 in evaluation of lupus Nephritis patients. Egypt Rheumatologist. 2022;44(2):151–7.

    Article  CAS  Google Scholar 

  65. Zhang T, Li H, Vanarsa K, Gidley G, Mok CC, Petri M, et al. Association of urine sCD163 with proliferative lupus Nephritis, fibrinoid necrosis, cellular crescents and intrarenal M2 macrophages. Front Immunol. 2020;11:671.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Nielsen AJ, Nielsen MC, Birn H, Ivarsen P, Møller HJ, Hviid CV. Urine soluble CD163 (sCD163) as biomarker in glomerulonephritis: stability, reference interval and diagnostic performance. Clin Chem Lab Med (CCLM). 2021;59(4):701–9.

    Article  CAS  PubMed  Google Scholar 

  67. Frantz C, Pezet S, Avouac J, Allanore Y. Soluble CD163 as a potential biomarker in systemic sclerosis. Disease markers. 2018;2018.

  68. Kowal-Bielecka O, Bielecki M, Guiducci S, Trzcinska-Butkiewicz B, Michalska-Jakubus M, Matucci-Cerinic M, et al. High serum sCD163/sTWEAK ratio is associated with lower risk of digital ulcers but more severe Skin Disease in patients with systemic sclerosis. Arthritis Res Therapy. 2013;15(3):1–11.

    Article  Google Scholar 

  69. Ding D, Song Y, Yao Y, Zhang S. Preoperative serum macrophage activated biomarkers soluble mannose receptor (sMR) and soluble haemoglobin scavenger receptor (sCD163), as novel markers for the diagnosis and prognosis of gastric cancer. Oncol Lett. 2017;14(3):2982–90.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Xia F, Qian C-R, Xun Z, Hamon Y, Sartre A-M, Formisano A et al. TCR and CD28 concomitant stimulation elicits a distinctive calcium response in naive T cells. Front Immunol. 2018:2864.

  71. Markovic-Plese S, Cortese I, Wandinger K-P, McFarland HF, Martin R. CD4 + CD28–costimulation-independent T cells in multiple sclerosis. J Clin Investig. 2001;108(8):1185–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Choi B-S, Park Y-K, Lee J-S. The CD28/HLA-DR expressions on CD4 + T but not CD8 + T cells are significant predictors for progression to AIDS. Clin Experimental Immunol. 2002;127(1):137–44.

    Article  CAS  Google Scholar 

  73. Mir M, Albaradie HA. R. Costimulation Immunotherapy against Transplantation, Autoimmunity and Lymphomas. India: Nova Science Publishers, Inc; 2013.

    Google Scholar 

  74. Esensten JH, Helou YA, Chopra G, Weiss A, Bluestone JA. CD28 costimulation: from mechanism to therapy. Immunity. 2016;44(5):973–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Evans EJ, Esnouf RM, Manso-Sancho R, Gilbert RJ, James JR, Yu C, et al. Crystal structure of a soluble CD28-Fab complex. Nat Immunol. 2005;6(3):271–9.

    Article  CAS  PubMed  Google Scholar 

  76. García-Chagollán M, Ledezma‐Lozano IY, Hernández‐Bello J, Sánchez‐Hernández PE, Gutiérrez‐Ureña SR, Muñoz‐Valle JF. Expression patterns of CD28 and CTLA‐4 in early, chronic, and untreated rheumatoid arthritis. J Clin Lab Anal. 2020;34(5):e23188.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Hakim M, Dror AF, Alishekevitz D, Meilin E, Shilovizky O, Weber JS, et al. CD28 shedding is a novel immune-regulatory mechanism found in cancer patients which directly inhibits anti PD-1 effect. Cancer Res. 2020;80(16Supplement):2846A–A.

    Article  Google Scholar 

  78. Hebbar M, Jeannin P, Magistrelli G, HATRON PY, Hachulla E, Devulder B, et al. Detection of circulating soluble CD28 in patients with systemic Lupus Erythematosus, primary Sjögren’s syndrome and systemic sclerosis. Clin Experimental Immunol. 2004;136(2):388–92.

    Article  CAS  Google Scholar 

  79. Brambila-Tapia AJL, Dávalos-Rodríguez IP, Gámez-Nava JI, González-López L, Medina-Díaz J, Bernard-Medina AG, et al. CD28 proximal promoter polymorphisms in systemic Lupus Erythematosus susceptibility. Rheumatol Int. 2012;32(7):2165–8.

    Article  CAS  PubMed  Google Scholar 

  80. Ledezma-Lozano I, Padilla-Martinez J, Leyva-Torres S, Parra-Rojas I, Ramirez-Duenas M, Pereira-Suárez AL, et al. Association of CD28 IVS3 + 17T/C polymorphism with soluble CD28 in rheumatoid arthritis. Dis Markers. 2011;30(1):25–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Cao J, Zou L, Chen P, Zhang L. Increased production of circulating soluble co-stimulatory molecules CTLA-4, CD28 and CD80 in patients with rheumatoid arthritis. Int Immunopharmacol. 2012;14(4):585–92.

    Article  CAS  PubMed  Google Scholar 

  82. Wong C, Lit L, Tam L, Li E, Lam C. Aberrant production of soluble costimulatory molecules CTLA-4, CD28, CD80 and CD86 in patients with systemic Lupus Erythematosus. Rheumatology. 2005;44(8):989–94.

    Article  CAS  PubMed  Google Scholar 

  83. Kakoulidou M, Pirskanen-Matell R, Lefvert A. Treatment of a patient with myasthenia gravis using antibodies against CD25. Acta Neurol Scand. 2008;117(3):211–6.

    Article  CAS  PubMed  Google Scholar 

  84. Wang H, Wang K, Zhong X, Dai Y, Wu A, Li Y, et al. Plasma sCD28, sCTLA-4 levels in neuromyelitis optica and multiple sclerosis during relapse. J Neuroimmunol. 2012;243(1–2):52–5.

    Article  CAS  PubMed  Google Scholar 

  85. Pyo JY, Yoon T, Ahn SS, Song JJ, Park Y-B, Lee S-W. Soluble immune checkpoint molecules in patients with antineutrophil cytoplasmic antibody-associated vasculitis. Sci Rep. 2022;12(1):1–9.

    Article  Google Scholar 

  86. Wong C, Lun S, Ko F, Ip W, Hui D, Lam C. Increased expression of plasma and cell surface co-stimulatory molecules CTLA-4, CD28 and CD86 in adult patients with allergic Asthma. Clin Experimental Immunol. 2005;141(1):122–9.

    Article  CAS  Google Scholar 

  87. Ip W, Wong C, Leung T, Lam C. Elevation of plasma soluble T cell costimulatory molecules CTLA-4, CD28 and CD80 in children with allergic Asthma. Int Arch Allergy Immunol. 2005;137(1):45–52.

    Article  CAS  PubMed  Google Scholar 

  88. Ip W, Wong C, Leung T, Lam C. Plasma concentrations of soluble CTLA-4, CD28, CD80 and CD86 costimulatory molecules reflect Disease severity of acute Asthma in children. Pediatr Pulmonol. 2006;41(7):674–82.

    Article  CAS  PubMed  Google Scholar 

  89. Chen H, Zhou J, Zhao X, Liu Q, Shao L, Zhu Y, et al. Characterization of multiple soluble immune checkpoints in individuals with different Mycobacterium tuberculosis Infection status and dynamic changes during anti-tuberculosis treatment. BMC Infect Dis. 2022;22(1):1–9.

    Article  Google Scholar 

  90. Cao J, Zhang L, Huang S, Chen P, Zou L, Chen H, et al. Aberrant production of soluble co-stimulatory molecules CTLA-4 and CD28 in patients with chronic Hepatitis B. Microb Pathog. 2011;51(4):262–7.

    Article  CAS  PubMed  Google Scholar 

  91. Dong MP, Hoang DV, Hai H, Hoang TH, Sato-Matsubara M, Hieu VN, et al. Soluble Immune checkpoint protein CD27 is a Novel Prognostic Biomarker of Hepatocellular Carcinoma Development in Hepatitis C virus–sustained virological response patients. Am J Pathol. 2022;192(10):1379–96.

    Article  CAS  PubMed  Google Scholar 

  92. Arikan S, Gümüş A, Küçükhüseyin Ö, Coşkun C, Turan S, Cacina C, et al. The effect of CTLA-4 and CD28 gene variants and circulating protein levels in patients with gastric cancer. Turkish J Biochem. 2017;42(5):551–8.

    Article  CAS  Google Scholar 

  93. Isitmangil G, Gurleyik G, Aker FV, Coskun C, Kucukhuseyin O, Arikan S, et al. Association of CTLA4 and CD28 gene variants and circulating levels of their proteins in patients with Breast cancer. in vivo. 2016;30(4):485–93.

    CAS  PubMed  Google Scholar 

  94. Rossi E, Zizzari IG, Di Filippo A, Acampora A, Pagliara MM, Sammarco MG et al. Circulating immune profile can predict survival of metastatic uveal Melanoma patients: results of an exploratory study. Hum Vaccines Immunotherapeutics. 2022:1–10.

  95. Wong C, Ho AW, Tong PC, Yeung C, Chan JC, Kong AP, et al. Aberrant expression of soluble co-stimulatory molecules and adhesion molecules in type 2 diabetic patients with Nephropathy. J Clin Immunol. 2008;28(1):36–43.

    Article  CAS  PubMed  Google Scholar 

  96. Fiorina P, Vergani A, Bassi R, Niewczas MA, Altintas MM, Pezzolesi MG, et al. Role of podocyte B7-1 in diabetic Nephropathy. J Am Soc Nephrol. 2014;25(7):1415–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Li Y, Jin L, Yan J, Zhang H, Zhang R, Hu C. CD28 genetic variants increase susceptibility to Diabetic Kidney Disease in Chinese patients with type 2 Diabetes: a cross-sectional Case Control Study. Mediators of inflammation. 2021;2021.

  98. Sakthivel P, Shively V, Kakoulidou M, Pearce W, Lefvert A. The soluble forms of CD28, CD86 and CTLA-4 constitute possible immunological markers in patients with abdominal aortic Aneurysm. J Intern Med. 2007;261(4):399–407.

    Article  CAS  PubMed  Google Scholar 

  99. Gu D, Ao X, Yang Y, Chen Z, Xu X. Soluble immune checkpoints in cancer: production, function and biological significance. J Immunother Cancer. 2018;6(1):1–14.

    Article  Google Scholar 

  100. Haile ST, Dalal SP, Clements V, Tamada K, Ostrand-Rosenberg S. Soluble CD80 restores T cell activation and overcomes Tumor cell programmed death ligand 1–mediated immune suppression. J Immunol. 2013;191(5):2829–36.

    Article  CAS  PubMed  Google Scholar 

  101. Haile ST, Horn LA, Ostrand-Rosenberg S. A Soluble Form of CD80 enhances Antitumor immunity by neutralizing programmed death Ligand-1 and simultaneously providing CostimulationCD80 costimulates and prevents PD-L1 immunosuppression. Cancer Immunol Res. 2014;2(7):610–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hogan J, Radhakrishnan J. Minimal change Disease in adults. Core concepts in parenchymal Kidney Disease. Springer; 2014. pp. 15–21.

  103. McHugh RS, Ratnoff WD, Gilmartin R, Sell KW, Selvaraj P. Detection of a soluble form of B7-1 (CD80) in synovial fluid from patients with arthritis using monoclonal antibodies against distinct epitopes of human B7-1. Clin Immunol Immunopathol. 1998;87(1):50–9.

    Article  CAS  PubMed  Google Scholar 

  104. Hock B, O’Donnell J, Taylor K, Steinkasserer A, McKenzie J, Rothwell A, et al. Levels of the soluble forms of CD80, CD86, and CD83 are elevated in the synovial fluid of rheumatoid arthritis patients. Tissue Antigens. 2006;67(1):57–60.

    Article  CAS  PubMed  Google Scholar 

  105. Ozkorucu D, Cetin N, Sav NM, Yildiz B. Urine and serum ghrelin, sCD80 and sCTLA-4 levels in doxorubicin-induced experimental Nephrotic Syndrome. Int Urol Nephrol. 2016;48(7):1187–96.

    Article  CAS  PubMed  Google Scholar 

  106. Cara-Fuentes G, Wasserfall CH, Wang H, Johnson RJ, Garin EH. Minimal change Disease: a dysregulation of the podocyte CD80–CTLA-4 axis? Pediatr Nephrol. 2014;29(12):2333–40.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Garin EH, Mu W, Arthur JM, Rivard CJ, Araya CE, Shimada M, et al. Urinary CD80 is elevated in minimal change Disease but not in focal segmental glomerulosclerosis. Kidney Int. 2010;78(3):296–302.

    Article  CAS  PubMed  Google Scholar 

  108. Fadriquela A, Kim C-S, Lee K-J, Kang SH, Kim MY, Lee J-H. Soluble type immune checkpoint regulators using multiplex luminex immunoassay in chronic Hepatitis B patients. J Clin Pathol. 2021;74(12):780–6.

    Article  CAS  PubMed  Google Scholar 

  109. Li W, Xia Y, Yang J, Guo H, Sun G, Sanyal AJ, et al. Immune checkpoint axes are dysregulated in patients with alcoholic hepatitis. Hepatol Commun. 2020;4(4):588–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Odagiri N, Hai H, Thuy LTT, Dong MP, Suoh M, Kotani K, et al. Early change in the plasma levels of circulating soluble immune checkpoint proteins in patients with unresectable hepatocellular carcinoma treated by lenvatinib or transcatheter arterial chemoembolization. Cancers. 2020;12(8):2045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Dong MP, Enomoto M, Thuy LTT, Hai H, Hieu VN, Hoang DV, et al. Clinical significance of circulating soluble immune checkpoint proteins in sorafenib-treated patients with advanced hepatocellular carcinoma. Sci Rep. 2020;10(1):1–10.

    Google Scholar 

  112. Hock B, Starling G, Patton W, Salm N, Bond K, McArthur L, et al. Identification of a circulating soluble form of CD80: levels in patients with hematological malignancies. Leuk Lymphoma. 2004;45(10):2111–8.

    Article  CAS  PubMed  Google Scholar 

  113. Biggar RJ, Christiansen M, Rostgaard K, Smedby KE, Adami HO, Glimelius B, et al. Immunoglobulin subclass levels in patients with non-hodgkin Lymphoma. Int J Cancer. 2009;124(11):2616–20.

    Article  CAS  PubMed  Google Scholar 

  114. Peng Y, Zhang C, Rui Z, Tang W, Xu Y, Tao X, et al. A comprehensive profiling of soluble immune checkpoints from the sera of patients with non-small cell Lung cancer. J Clin Lab Anal. 2022;36(2):e24224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Wang Q, He Y, Li W, Xu X, Hu Q, Bian Z et al. Soluble immune checkpoint-related proteins in blood are associated with invasion and progression in non-small cell Lung cancer. Front Immunol. 2022;13.

  116. Simone R, Saverino D. The soluble CTLA-4 receptor and its emerging role in autoimmune Diseases. Curr Immunol Rev. 2009;5(1):54–68.

    Article  CAS  Google Scholar 

  117. Wang XB, Giscombe R, Yan Z, Heiden T, Xu D, Lefvert A. Expression of CTLA-4 by human monocytes. Scand J Immunol. 2002;55(1):53–60.

    Article  CAS  PubMed  Google Scholar 

  118. Pawlak E, Kochanowska IE, Frydecka I, Kielbinski M, Potoczek S, Bilinska M. The soluble CTLA-4 receptor: a new marker in autoimmune diseases. ARCHIVUM IMMUNOLOGIAE ET THERAPIAE EXPERIMENTALIS-ENGLISH EDITION-. 2005;53(4):336.

  119. Ward FJ, Dahal LN, Wijesekera SK, Abdul-Jawad SK, Kaewarpai T, Xu H, et al. The soluble isoform of CTLA‐4 as a regulator of T‐cell responses. Eur J Immunol. 2013;43(5):1274–85.

    Article  CAS  PubMed  Google Scholar 

  120. Saverino D, Simone R, Bagnasco M, Pesce G. The soluble CTLA-4 receptor and its role in autoimmune Diseases: an update. Autoimmun Highlights. 2010;1(2):73–81.

    Article  CAS  Google Scholar 

  121. Sakthivel P, Wermeling F, Elmgren A, Hulthe J, Kakoulidou M, Kari Lefvert A, et al. Circulating soluble CTLA-4 is related to inflammatory markers in the 70 year old population. Scand J Clin Lab Investig. 2010;70(4):237–43.

    Article  CAS  Google Scholar 

  122. Liu MF, Wang CR, Chen PC, Fung LL. Increased expression of soluble cytotoxic T-lymphocyte‐associated antigen‐4 molecule in patients with systemic Lupus Erythematosus. Scand J Immunol. 2003;57(6):568–72.

    Article  CAS  PubMed  Google Scholar 

  123. Dahal LN, Basu N, Youssef H, Khanolkar RC, Barker RN, Erwig LP, et al. Immunoregulatory soluble CTLA-4 modifies effector T-cell responses in systemic Lupus Erythematosus. Arthritis Res Therapy. 2016;18(1):1–11.

    Article  Google Scholar 

  124. Toussirot É, Saas P, Deschamps M, Pouthier F, Perrot L, Perruche S, et al. Increased production of soluble CTLA-4 in patients with spondylarthropathies correlates with Disease activity. Arthritis Res Therapy. 2009;11(4):1–11.

    Article  Google Scholar 

  125. Saverino D, Brizzolara R, Simone R, Chiappori A, Milintenda-Floriani F, Pesce G, et al. Soluble CTLA-4 in autoimmune thyroid Diseases: relationship with clinical status and possible role in the immune response dysregulation. Clin Immunol. 2007;123(2):190–8.

    Article  CAS  PubMed  Google Scholar 

  126. Oaks MK, Hallett KM. Cutting edge: a soluble form of CTLA-4 in patients with autoimmune thyroid Disease. J Immunol. 2000;164(10):5015–8.

    Article  CAS  PubMed  Google Scholar 

  127. Daroszewski J, Pawlak E, Karabon L, Frydecka I, Jonkisz A, Slowik M, et al. Soluble CTLA-4 receptor an immunological marker of Graves’ Disease and severity of ophthalmopathy is associated with CTLA-4 Jo31 and CT60 gene polymorphisms. Eur J Endocrinol. 2009;161(5):787–93.

    Article  CAS  PubMed  Google Scholar 

  128. Sato S, Fujimoto M, Hasegawa M, Komura K, Yanaba K, Hayakawa I, et al. Serum soluble CTLA-4 levels are increased in diffuse cutaneous systemic sclerosis. Rheumatology. 2004;43(10):1261–6.

    Article  CAS  PubMed  Google Scholar 

  129. Łuszczek W, Kubicka W, Jasek M, Baran E, Cisło M, Nockowski P, et al. CTLA-4 gene polymorphisms and natural soluble CTLA‐4 protein in psoriasis vulgaris. Int J Immunogenet. 2006;33(3):217–24.

    Article  PubMed  Google Scholar 

  130. Simone R, Brizzolara R, Chiappori A, Milintenda-Floriani F, Natale C, Greco L, et al. A functional soluble form of CTLA-4 is present in the serum of celiac patients and correlates with mucosal injury. Int Immunol. 2009;21(9):1037–45.

    Article  CAS  PubMed  Google Scholar 

  131. AlFadhli S. Overexpression and secretion of the soluble CTLA-4 splice variant in various autoimmune Diseases and in cases with overlapping autoimmunity. Genetic Test Mol Biomarkers. 2013;17(4):336–41.

    Article  CAS  Google Scholar 

  132. Simone R, Pesce G, Antola P, Rumbullaku M, Bagnasco M, Bizzaro N et al. The soluble form of CTLA-4 from serum of patients with autoimmune diseases regulates T-cell responses. BioMed research international. 2014;2014.

  133. Shi H-Z, Mo X-Y, Zhong X-N. Soluble CTLA-4 in sera of patients with bronchial Asthma. J Asthma. 2005;42(2):133–9.

    Article  CAS  PubMed  Google Scholar 

  134. Santoso B, Sa’adi A, Dwiningsih SR, Tunjungseto A, Widyanugraha MYA, Mufid AF, et al. Soluble immune checkpoints CTLA-4, HLA‐G, PD‐1, and PD‐L1 are associated with endometriosis‐related infertility. Am J Reprod Immunol. 2020;84(4):e13296.

    Article  CAS  PubMed  Google Scholar 

  135. Simone R, Tenca C, Fais F, Luciani M, De Rossi G, Pesce G et al. A soluble form of CTLA-4 is present in paediatric patients with acute lymphoblastic Leukaemia and correlates with CD1d + expression. 2012.

  136. Pistillo MP, Fontana V, Morabito A, Dozin B, Laurent S, Carosio R, et al. Soluble CTLA-4 as a favorable predictive biomarker in metastatic Melanoma patients treated with ipilimumab: an Italian Melanoma intergroup study. Cancer Immunol Immunother. 2019;68(1):97–107.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Taylor and Francis Publisher for letting us reuse figure (Fig. 1) in this article.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Andrea Aprilia wrote the main manuscript text, made Fig. 2, and asked copyright for Fig. 1. Kusworini Handono, Hidayat Sujuti, Akhmad Sabarudin, and Nuning Winaris supervised and gave suggestions to make the review article suitable to be published. Nuning Winaris proofreads the language of the manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Kusworini Handono.

Ethics declarations

Conflict of interest

All authors declare no conflict of interest.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aprilia, A., Handono, K., Sujuti, H. et al. sCD163, sCD28, sCD80, and sCTLA-4 as soluble marker candidates for detecting immunosenescence. Immun Ageing 21, 9 (2024). https://doi.org/10.1186/s12979-023-00405-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12979-023-00405-0

Keywords