Next Article in Journal
Unraveling the Potential Role of Tecomella undulata in Experimental NASH
Next Article in Special Issue
Pathways of Coagulopathy and Inflammatory Response in SARS-CoV-2 Infection among Type 2 Diabetic Patients
Previous Article in Journal
Cell Sorting-Directed Selection of Bacterial Cells in Bigger Sizes Analyzed by Imaging Flow Cytometry during Experimental Evolution
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19

1
West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
2
Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BE, UK
3
State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
4
Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610041, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(4), 3246; https://doi.org/10.3390/ijms24043246
Submission received: 22 December 2022 / Revised: 1 February 2023 / Accepted: 3 February 2023 / Published: 7 February 2023
(This article belongs to the Special Issue COVID-19 Coagulopathy: Advances on Pathophysiology and Therapies)

Abstract

:
Acute pancreatitis is a common gastrointestinal disease with increasing incidence worldwide. COVID-19 is a potentially life-threatening contagious disease spread throughout the world, caused by severe acute respiratory syndrome coronavirus 2. More severe forms of both diseases exhibit commonalities with dysregulated immune responses resulting in amplified inflammation and susceptibility to infection. Human leucocyte antigen (HLA)-DR, expressed on antigen-presenting cells, acts as an indicator of immune function. Research advances have highlighted the predictive values of monocytic HLA-DR (mHLA-DR) expression for disease severity and infectious complications in both acute pancreatitis and COVID-19 patients. While the regulatory mechanism of altered mHLA-DR expression remains unclear, HLA-DR−/low monocytic myeloid-derived suppressor cells are potent drivers of immunosuppression and poor outcomes in these diseases. Future studies with mHLA-DR-guided enrollment or targeted immunotherapy are warranted in more severe cases of patients with acute pancreatitis and COVID-19.

1. Introduction

New insights into the mechanisms of pathology can sometimes arise from similarities between fundamentally different diseases. This effect can be most pronounced during the emergence of a new infectious disease, such as the recent COVID-19 pandemic. One such unlikely pairing is acute pancreatitis (AP) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
AP is a sterile inflammatory disorder of the pancreas with an increasing global incidence [1] affecting around 2.8 million patients annually [2]. The etiology of AP is diverse and includes gallstones, alcohol excess, hypertriglyceridemia, endoscopic retrograde cholangiopancreatography, certain medicines, and other rarer causes [3]. Most cases of AP patients are mild and uneventful given that supportive care is in time and appropriate. However, some are more severe, which involve local complications (acute pancreatic necrosis or fluid collection; moderately severe acute pancreatitis, MSAP) and/or persistent organ failure (SOFA score of respiratory, circulatory, and renal system equal or more than 2 lasting > 48 h; severe acute pancreatitis, SAP) [4]. Feed-forward auto-amplification of the initial cellular injury in SAP [5,6] results in persistent systemic inflammatory response syndrome (SIRS), multiple organ dysfunction syndrome (MODS), infection, and death. Persistent organ failure [7,8,9,10] and infected pancreatic necrosis [11,12], alone or in combination, are key determinants of severity in AP and contribute to an immune anergy, secondary infections, and a mortality of > 30%. Currently, there are no specific therapies effectively targeting the initial cellular injury or determinants that resulting in MODS [13].
COVID-19, on the other hand, is a potentially lethal infectious disease caused by the enveloped, positive-strand RNA, SARS-CoV-2, affecting over 600 million cases globally [14]. The disease spectrum of COVID-19 is also highly variable, ranging from asymptomatic (test-positive) disease to critical illness (respiratory failure, septic shock, and/or MODS) [15,16]. SARS-CoV-2 mainly utilizes the angiotensin-converting enzyme 2 (ACE2) as the human host cell entry receptor [17], which is ubiquitously expressed in the nasal epithelium, lung, heart, intestine, and kidney and rarely expressed on immune cells [18]. ACE2 is also expressed on pancreatic ductal cells, acinar cells, and islet cells, making the pancreas vulnerable to viral infection [19]. Serum pancreatic enzymes are elevated in 25% of patients suffering COVID-19, which is linked to worsened clinical outcomes including mechanical ventilation and mortality even in those without AP [20,21,22]. Patients with COVID-19 who developed AP during hospitalization also have a more severe clinical course [23], and indeed SARS-CoV-2 may itself precipitate an episode of AP with marked metabolic derangement even in the absence of local complications or organ failure [24]. More importantly, however, patients with severe/critical COVID-19 appear to be increasingly susceptible to secondary infections [25,26] as a result of immune anergy in a similar manner to SAP.
Dysregulated immune responses in SAP and severe/critical COVID-19 have similar patterns of cytokine release and share many pathways of cellular immunity, especially immunosuppression-related monocyte deactivation in the form of downregulated expression of monocytic human leukocyte antigen-DR (HLA-DR) [27,28]. This review summarizes the role of monocytic HLA-DR (mHLA-DR) expression in the development of immunosuppression and organ failure in both SAP and severe/critical COVID-19.

2. Pathogenesis and Immunopathology in AP and COVID-19

2.1. Pathophysiological Mechanisms in AP and COVID-19

Diverse stimuli evoke inflammatory cascades with apparently analogous patterns and clinical manifestations, implying similarities in the pathogenesis and symptomatology of AP and COVID-19 [29]. Cytokines and damage-associated molecular patterns (DAMPs), such as histones, high-mobility group box-1 protein, hyaluronan fragments, mitochondrial DNA, and heat-shock proteins are released from dying or injured cells in the injured pancreas or SARS-CoV-2 infected tissues—particularly lungs. This is associated with and results from a series of molecular events, including premature trypsinogen activation, calcium overload, mitochondria failure, endoplasmic reticulum stress, impaired autophagy, or by SARS-CoV-2 proliferation and release, respectively [6,30,31,32,33]. Interaction of DAMPs with pattern-recognition receptors (PRRs), including Toll-like receptors and NLRP3 inflammasome of the adjacent parenchymal cells or immune cells, promotes the production of various pro-inflammatory cytokines and chemokines [31,34,35,36]. Of note, cell death pathways (e.g., autophagy, NETosis, pyroptosis, apoptosis, necroptosis, and ferroptosis) in surrounding immune cells and stromal cells are activated, fueling the cytokine storm and cultivating a positive cell death-inflammation feedback loop [30,37,38]. In COVID-19, virus particles themselves act as pathogen-associated molecular patterns (PAMPs), which could also be identified by PRRs and activate local inflammation and an innate immune response, evoking the cytokine storm and assembling those induced by DAMPs [29,39]. Activated circulating leukocytes, particularly monocytes, are then recruited to the inflamed pancreas or infected lungs, provoking systemic inflammation and organ failure in AP and COVID-19 alike [29,40,41,42,43]. Moreover, monocytes/macrophages could be infected by SARS-CoV-2, triggering massive inflammatory responses in COVID-19 [44].
The involvement of adaptive immunity in AP has been recognized, but its precise role in the sterile inflammatory response seen in AP remains poorly characterized [45]. In contrast, SARS-CoV-2 directly activates specific T cell subsets, initiating an adaptive immune response [46]. Persistent viral stimulation, however, leads to T cell exhaustion, with reduced effector functions and proliferative capacity [47]. This T cell exhaustion phenomenon can also be observed in AP patients [48].
Levels of several circulating pro-inflammatory cytokines are dramatically elevated and closely correlate with the development of SAP or severe/critical COVID-19 [49,50,51,52]. Patterns of cytokine alterations in AP and COVID-19 were shown to be remarkably similar in a recent meta-analysis, with tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-8, and IL-10 concentrations significantly higher in more severe forms than non-severe forms of the two diseases [53]. The crosstalk between excessive inflammatory cytokines, platelet activation, complement activation, and endothelial injury forms a deleterious hyper-inflammatory and hyper-coagulopathy environment which is associated with life-threatening complications (i.e., coagulopathy and vascular immune-thrombosis) of AP and COVID-19 [51,54,55,56,57,58].
Systemic lipotoxicity deserves to be highlighted in this context. In severe/critical COVID-19, lipotoxicity can trigger multiple organ failure and mortality resembling SAP [59]. SARS-CoV-2 can directly infect adipose tissue and promotes the release of several inflammatory cytokines [60]. The pancreas itself is a target of SARS-CoV-2, resulting in the interstitial leakage of pancreatic lipase which induces lipolysis of intrapancreatic adipose tissue and release of excess unsaturated fatty acids (UFAs). These toxic UFAs in turn further directly lead to parenchymal cell injury and provoke the release of pro-inflammatory mediators, driving the cytokine storm and organ failure in SAP and severe/critical COVID-19 [59,61,62]. Lipase inhibitors have been shown to ameliorate lipolysis-induced cytokine storms and mortality [61,62,63,64].
In summary, the pathophysiological mechanisms of AP and COVID-19 share many similarities including cell death-inflammation cascade, cytokine storms, enhanced lipolysis, and dysregulated immune responses. These immune responses will be discussed in the next section.

2.2. Altered Immune Responses in AP and COVID-19

Immune anergy, evidenced by the failure of delayed hypersensitivity responses, correlates with the development of sepsis and mortality in trauma and surgical patients [65,66,67], as well as in SAP [68]. In the first stage of SAP, an excessive pro-inflammatory burst is rapidly followed by an anti-inflammatory reaction that may result in a generalized inflammatory response in sites remote from the initial pancreatic injury site and gives rise to SIRS [69,70,71]. There is a compensatory response to counteract the overwhelming pro-inflammatory state [72], which may ultimately result in immune suppression [73]. In 1996, Bone termed this immunological phenomenon as “compensatory anti-inflammatory response syndrome” (CARS) [65,66,72].
Unlike SIRS, which is clearly defined by clinical parameters, CARS lacks clinical manifestations and can only be defined molecularly by a combination of immunological alterations. In the landmark paper of Volk’s group in 1997, it was described that many septic patients who died from nosocomial infections had associated downregulation of mHLA-DR [74]. Monocytes from these patients had reduced capacity to act in a pro-inflammatory manner by producing TNF-α following stimulation of lipopolysaccharide (LPS) in vitro, termed “immunoparalysis” [74,75]. Where CARS was once thought to follow sequentially from SIRS, current thinking views CARS responses as concomitant to SIRS; balance in both responses restores homeostasis, but an overshoot of the mechanisms of either SIRS or CARS leads to further injury by excessive inflammation or secondary infection and, ultimately, organ failure and death [67,76,77,78,79,80,81,82,83]. Development of CARS results in lymphocyte apoptosis, T lymphocyte anergy, and deactivation of monocytes resulting in reduced mHLA-DR expression. Furthermore, CARS is associated with elevated levels of circulating IL-10, transforming growth factor-beta (TGF-β) and other anti-inflammatory cytokines, which contribute to the risk of secondary infection.
Immune response to SARS-CoV-2 is characterized by the failure of robust type I and type III interferon response and high expression of pro-inflammatory cytokines and chemokines [17]. Like AP, immune alterations, including severe lymphopenia and functional monocyte deactivation, are indicative of immunosuppression in severe/critical COVID-19 patients [84]. Indeed, monocytes exhibit heterogeneous, dynamic, and severity-dependent alterations of transcription and immune phenotype upon acute pathological insults which appear similar in both SAP and severe/critical COVID-19 patients (Figure 1).
Inflammatory monocytes are enriched in the lungs of severe/critical COVID-19 patients and are also the most altered pancreatic immune cells during progression and recovery of AP [85,86]. Decreased monocytic expression of HLA-DR has a predictive value for the poor prognosis of patients with sepsis [87,88], and the level of mHLA-DR expression may identify patients who are susceptible to the development of infectious complications after trauma [89], major surgery [90], and burns [91]. Here, we review the utility of mHLA-DR in assessing the state of the immune response in AP and COVID-19 and detail-relevant implications for therapy.

3. Structure and Expression of mHLA-DR

HLA-DR is a type of major histocompatibility complex (MHC) II molecule [92]. It is a heterodimeric glycoprotein composed of the 33–35 kD heavy/α chain and the 27–29 kD light/β chain, assembling into a structure comprising a peptide binding site on top of two immunoglobulin domains [92]. Encoded by adjacent genes, the β chain is polymorphic around the amino acid residues of the peptide-binding site in contrast to the invariant α chain [93].
HLA-DR is mostly expressed on antigen-presenting cells (APCs) such as monocytes, macrophages, dendritic cells, and B cells. The primary function of HLA-DR is to present peptide antigens to the immune system for the purpose of eliciting or suppressing T-(helper)-cell responses, eventually leading to the production of antibodies against the same peptide antigen. HLA-D/DR-controlled antigens play an essential role in the cell-to-cell interactions required to generate an immune response [94,95].
The biosynthesis, trafficking, and recycling of HLA-DR are regulated by multiple factors affecting cell surface expression. Consequently, the tightly regulated level of HLA-DR expression on the surface of monocytes is thought to be an indicator for monocyte function and the state of the immune response, with high levels of mHLA-DR associated with enhanced antigen presenting capacity and immune activation, and low levels associated with immune suppression.

3.1. Measurement of mHLA-DR

Several reviews [67,96,97] have emphasized the importance of flow cytometry as an indicator of immune function in clinical practice. The unit of measurement of HLA-DR via flow cytometer can be the percentage of HLA-DR positive monocytes (%), the mean fluorescence intensity (MFI), the fluorescence unit relative to the monocyte population (RFU), or antibodies per cell (AB/c). Due to the dynamic nature of HLA-DR expression and recycling, it is critical that measurement of expression is standardized. We support the process published by Docke’s and Monnaret’s groups [98,99,100], which have been widely tested and published and appear to result in a strong correlation between transcription and cell surface expression of mHLA-DR. It should be highlighted that a percentage of HLA-DR+ monocytes less than 30% or values of AB/c below 5000 represents immunoparalysis, and values greater than 80% or 15 000 AB/c indicate immunocompetence [99]. The critical features for the sampling and measurement of mHLA-DR from human plasma samples are summarized in Figure 2.

3.2. Regulation of mHLA-DR Expression

The transcription of mHLA-DR is complex and heterogeneous, mediated by a series of conserved cis-acting regulatory promoter elements and interacting transcription factors [102]. Among these, class II transactivator (CIITA) is the master regulator of HLA-DR transcription [103]. Polymorphisms of CIITA promoter are associated with decreased mHLA-DR expression in patients with septic shock [104]. Besides biosynthesis, the expression of mHLA-DR can be post-translationally regulated by exocytosis, stability, and recycling. The class II-associated Ii peptide (CLIP), generated from cleavage of CD74 (MHC class II invariant chain, Ii) via members of the cathepsin family, is critical for the transport of HLA-DR to the cell surface [105]. In CD74 knockout mice, MHC II molecules are mainly retained in endoplasmic reticulum with reduced levels on the cell surface [106]. Reducing CLIP generation by blocking cysteine protease activity reduced surface MHC II expression, including HLA-DR to 60% on human monocytes in steady state [107]. HLA-DM, the key accessory molecules in the MHC class II loading compartment, catalyzes the dissociation of CLIP in exchange for more stably binding peptides [108]. MHC II molecules on the cell surface are normal in amounts but mainly loaded with CLIP in HLA-DM-deficient mice [109]. HLA-DR loaded with high-affinity peptides are postulated to be more stable than those with CLIP, indicating the role of HLA-DM in regulating mHLA-DR expression [107]. Of note, surface HLA-DR could be internalized, exchanged from lower affinity peptides into other peptides, and rapidly recycled back to the cell surface [110]. In summary, expression of mHLA-DR is finely regulated by multiple steps, including biosynthesis, peptide-loading via cathepsin-induced CLIP and HLA-DM, vesicular transport to the cell surface, and recycling (Figure 3).
Multiple pro- and anti-inflammatory cytokines are reported to dynamically control the expression of mHLA-DR [112]. The main mechanisms of cytokines modulating HLA-DR expression are summarized in Table 1. However, the detailed regulatory mechanisms of various cytokines on mHLA-DR expression remain largely unknown.

4. The Role of mHLA-DR in AP and COVID-19

Monocytic HLA-DR expression alters dynamically in response to the variation of immune responses in the body during the disease course of AP and COVID-19. Evaluating the dynamic expression of mHLA-DR provides indicative information for diagnosis and prediction of disease severity, infectious complications, and prognosis (Figure 4).
The expression of mHLA-DR on admission was downregulated in AP patients compared to healthy controls; it further decreased on days 1 and 2 with differential degrees depending on severity [123,124,125]. While mHLA-DR expression recovered rapidly at day 3 and became normal after day 7 in less severe patients, it persisted at low levels for 1–2 weeks in more severe cases [124,126]. Indeed, mHLA-DR expression displays an inverse relationship with severity throughout at least the first three weeks of disease [127], with the lowest expression of mHLA-DR in SAP consistently recorded between 48 and 72 h of disease onset [127,128].
Overall, mHLA-DR expression either increases or decreases slightly in mild COVID-19 patients compared with healthy controls [129,130]. However, a marked and persistent decrease in expression is described in severe/critical COVID-19 patients in most studies [129,131,132,133,134,135,136,137,138,139,140]. The immune response to severe COVID-19 can be categorized into three groups according to the kinetics of mHLA-DR expression: (i) hyperactivated monocytes/macrophage phenotype (persistently high mHLA-DR > 30,000 AB/c)—strongly associated with mortality; (ii) prolonged immunodepression (persistently low mHLA-DR < 15,000 AB/c after days 5–7)—strongly correlating with secondary infection; (iii) transient immunodepression (early mHLA-DR < 15,000 AB/c, rising above 15,000 AB/c after 5–7 days)—at risk of secondary infection [141]. Patients with acute respiratory distress syndrome (ARDS) secondary to COVID-19 exhibit either immune dysregulation evidenced by very low mHLA-DR expression (i.e., lower than 5000 AB/c) and depletion of lymphocytes, or macrophage activation syndrome characterized by elevated ferritin, where associated HLA-DR levels might be reduced [142], or comparable to healthy controls [143]. Expression of mHLA-DR may be able to provide some information on disease course and has been observed to normalize upon recovery from critical illness in patients with COVID-19 (from 1–3 days to over 10 days after admission), but continued to fall in a patient who died [136]. Critically ill COVID-19 patients with long hospital stays (>25 days) presented with a more profound reduction in mHLA-DR expression than patients with short hospital stays (<25 days) [140]. Furthermore, convalescent COVID-19 patients exhibit mHLA-DR levels which are higher than those of healthy controls at 6 months, and equal to healthy controls at 9 and 12 months following discharge from the hospital [140,144].
The reduction of HLA-DR expression in COVID-19 patients has been reported in both classical monocytes [144,145], as well as intermediate monocytes and/or non-classical monocytes [132,146,147], although usually in one group or the other, depending on the respective study. Classical monocytes are the first peripheral immune cell type to recover HLA-DR positivity during the recovery of critically ill COVID-19 patients [148].

4.1. Severity Prediction Using mHLA-DR in AP and COVID-19

The predictive values of mHLA-DR for severity and mortality of AP and COVID-19 are summarized in Table 2.
MHLA-DR expression is inversely correlated with surrogate biochemical markers of severity (C-reactive protein [CRP], TNF-α, and IL-6) [127,128,154,155,156,157], clinical scoring systems (Ranson, Acute Physiology and Chronic Health Evaluation II [APACHE II], and MODS criteria) [128,154,157,158], and actual severity of AP [125,127,128,150,151,156,158,159,160], and AP patients with low mHLA-DR expression had approximately 2.7 times longer ICU stays than those with normal expression [159]. HLA-DR expressed on classical monocytes was able to distinguish cases of mild from MSAP/SAP and SAP from MSAP on admission [160]. Indeed, mHLA-DR expression on admission, days 2 and 5 all have been shown to have predictive value for SAP [150] and/or the subsequent development of organ failure(s) [151].
The utility of mHLA-DR to predict mortality in AP is more controversial. While several studies have reported differences in mHLA-DR expression between survivors and non-survivors on days 7 [124,128] or 10 after admission [152], others found no difference [127,158,161]. These results might be explained by the differences in the design of the respective clinical studies, or by the heterogeneous and dynamic immune response in the study populations.
Despite one study finding that mHLA-DR expression was irrelevant to severity of COVID-19 [146], most studies demonstrate an inverse relationship [135,136,137,139]. Low or very low mHLA-DR expression has been described in association with ARDS [162], severe respiratory failure [142], thrombocytopenia, increased antibiotic requirements, and need for extracorporeal membrane oxygenation [134,135,142,153,162]. Similarly to AP, lower levels of mHLA-DR expression correlated with length of hospital stay [140], SOFA score [153], and serum clinical biochemical parameters including D-dimer, lactate dehydrogenase, CRP, procalcitonin, ferritin, IL-6, IL-10, granulocyte colony-stimulating factor, chemokine C-X-C motif ligand 10, chemokine C-C motif ligand 2 (CCL2), and IFN-γ levels [135,136,137,144,147,153,163]. Although overall mHLA-DR expression does not appear to differ between survivors and non-survivors [141], the lowest levels of mHLA-DR expression can be observed in patients with COVID-19 who died in the ICU [84]. The proportion of mHLA-DR+ monocytes was also lower in deceased COVID-19 patients compared with time-matched controls [164]. Expression of mHLA-DR recovers with clinical improvement but continues to fall in patients who do not survive [134,144].

4.2. Prediction of Infectious Complications using mHLA-DR

MHLA-DR regulates the interplay between innate and adaptive immunity and represents an overview of an organism’s capacity for antigen presentation, cytokine production, and phagocytosis [165]. HLA-DR downregulation is not only limited to the blood compartment but can also be observed in lymphatic tissue [166]. With standardization of flow cytometry-based measurement of mHLA-DR, a multicenter comparison of obtained results becomes feasible [98]. Therefore, mHLA-DR is now the most frequently utilized biomarker for assessing the development of immunosuppression in critically ill patients, including sepsis, stroke, trauma, and burns [167]. Following the rationale of immunosuppression in severe critical illnesses, mHLA-DR expression levels are predictive of septic complications in AP and COVID-19 using values shown in Table 3.
Failure of the intestinal barrier function is often thought to be responsible for the dysregulated systemic inflammation in AP [126,168]. The proportion of HLA-DR+ monocytes correlated negatively with measures of small intestinal permeability, including the urinary lactulose/mannitol ratio and D(-)-lactate concentrations [126]. AP patients with infectious complications, including sepsis or infected pancreatic necrosis, had lower HLA-DR expression which recovered at a slower rate than those without [127,128,150,152,154,157,169,170,171]. The relative risk of developing infected pancreatic necrosis in AP patients with low mHLA-DR expression that persisted into the second week of illness was 11.3 (1.6–82.4) [170], and persistently low HLA-DR levels have even been shown to be related to multidrug resistant infection [171]. This ability to identify patients with infectious complications early was as good or superior to routine biochemical markers and clinical scoring systems including CRP and APACHE II [152]. Therefore, a persistently low expression of mHLA-DR might be an effective and reliable indicator of potentially lethal infectious complications in patients with AP that could perhaps be used to identify patients who might benefit from early antimicrobial therapy.
As in AP, persistently low levels of mHLA-DR expression are associated with secondary infection in COVID-19 patients [172]. COVID-19 patients who developed secondary bacterial infections exhibit lower levels of mHLA-DR expression than those without at all time points (days 1, 4, and 7 [173], days 5–7, days 8–10 [141]). MHLA-DR expression (AB/c) on days 0–3 and on days 7–10 have been shown to predict secondary infection in COVID-19 patients in the ICU [134].

4.3. Regulation of mHLA-DR Expression in AP and COVID-19

Both pro- and anti-inflammatory cytokines, including TNF-α, IL-6, IL-8, IL-10, and IL-1RA1, can downregulate—and correlate inversely with—mHLA-DR expression in AP patients [156,160]. IL-6, IL-8, IL-10, and IL-1RA1 inhibit HLA-DR expression on classical monocytes in vitro [160]. TNF-α enhances IL-10 production of monocytes in vitro and downregulates levels of HLA-DR, even in the presence of anti-IL-10 monoclonal antibodies, demonstrating inhibition of mHLA-DR expression via an alternate pathway [156].
The regulatory mechanisms of reduced mHLA-DR expression in severe/critical COVID-19 patients are less well understood, but IL-6 and IL-10 are similarly thought to be possible drivers to reduce mHLA-DR expression in the disease. MHLA-DR expression was strongly reduced by plasma from COVID-19 patients with immune dysregulation but not healthy controls [130,142]; the effect could be partially restored by the addition of the IL-6 blocker Tocilizumab [142]. The highly expressed cytokines in COVID-19 patients included IL-10, IL-6, IL-7, TNF-α, IFN-α, CCL2, and CCL4, but only incubation monocytes with IL-10 downregulated HLA-DR expression [130].
The altered cytokine profiles in sterile or infectious inflammatory diseases including AP and COVID-19 are dynamic and complex, which may affect mHLA-DR expression synergistically or antagonistically. Future studies are needed to investigate the precise role of cytokines in regulating mHLA-DR so as to develop potential therapeutic targets in immune regulation.

4.4. Monocytic Myeloid-Derived Suppressor Cells

A proportion of circulating HLA-DR−/low monocytes seen in both AP and COVID-19 patients have been identified as CD14+CD11b+HLA-DR−/lowCD15 monocytic myeloid-derived suppressor cells (M-MDSCs); these cell types may cloud earlier studies on the topic, as they could be misidentified as HLA-DR−/low classical monocytes [174,175,176]. M-MDSCs are characterized by their potent immunosuppressive effects on other immune cells, especially T cells, through various mechanisms including secretion of arginase-1 (Arg-1), and inducible nitric oxide synthase, production of reactive oxygen and nitrogen species, secretion of cytokines including TGF-β and IL-10, and induction of regulatory T cells [175].
The proportion of M-MDSCs in peripheral blood mononuclear cells correlates with AP severity as reflected by plasma CRP levels, APACHE II score, and length of stay [174]. Increased levels of Arg-1 and ROS can further be observed in AP patients, especially those with a severe clinical course [174]. Similarly, expansion of M-MDSCs was reported together with increased Arg-1 activity in plasma, and these are associated with severity and fatal outcome in COVID-19 patients [175].
Therapeutic approaches aimed at reducing the number, function, and accumulation of M-MDSCs might improve the suppressive state of the immune system and improve complication-free survival in both SAP and severe/critical COVID-19 patients.

5. Conclusions and Future Prospects

MHLA-DR expression serves as a useful biomarker for immune (dys)function in patients with AP and COVID-19. The measurement of patterns and dynamics of mHLA-DR expression in both these diseases can help clinicians to determine the severity and prognosis, and perhaps guide timing and selection of therapy. Monitoring mHLA-DR expression appears to help identify and differentiate patients at higher risk of secondary infections associated with poor outcomes. While immunosuppression in general is thought to represent later stages of both diseases, in actual fact, time course and immune responses can be highly heterogeneous and variable [127,177]. MHLA-DR modulation occurs over several days [178], necessitating multiple, consecutive mHLA-DR measurements following a standardized assessment procedure of flow cytometry in patients from point of admission. MHLA-DR measurement should be prioritized for patients with clinically severe presentations with rapidly worsening organ dysfunctions or who are in need of invasive treatments or are at high risk of infectious complications with poor prognosis [179,180,181].
Examples of potential mHLA-DR-directed interventions that could find utility in AP and COVID-19 include several immunostimulatory agents, including IFN-γ [155,182,183,184], recombinant IL-7 [185], and granulocyte-macrophage colony-stimulating factor [155]. Thymosin alpha 1 (Tα1), a peptide hormone used to stimulate the T-cell mediated immune response, has been tested in patients with predicted for necrotizing pancreatitis (presumably immunocompromised), but results are so far disappointing [186]; thus far, there has been no demonstrable reduction in the incidence of infected pancreatic necrosis, new-onset organ failure, or any other complications. Defining immunosuppression, for example, by using the measurement of mHLA-DR expression to guide participant selection and/or tailor the treatment dose, may be required to demonstrate effective immune-stimulatory therapy.
The complex and highly variable immune alterations seen in severe acute inflammation and infection warrant stratified immunotherapy. MHLA-DR expression provides supportive information in determining the timing and strategies of individual immune treatments, including anti-inflammatory, immune-stimulatory or immune-modulatory agents at different disease stages, something that has been demonstrated in both acute pancreatitis and COVID-19. The emergence of a new global pandemic disease has provided valuable insights into the mechanisms of a long-established illness, with considerable potential to draw insights into one disease from the other. There is a need for a simple, cheap, and effective universal immune assessment tool, combining mHLA-DR with established clinical markers of disease severity and possibly other circulating immune cell profiles to aid assessment of the disease course of illnesses with a systemic inflammatory component in order to predict outcomes and to guide treatment decisions.

Author Contributions

Conceptualization, S.L., W.H.; investigation, S.L., W.L.; writing original draft preparation, S.L.; writing-review and editing, S.L., W.L., P.S., X.Z., W.H.; visualization, S.L., W.L., R.S., W.H.; supervision, P.S., X.Z., J.-W.L., L.C., D.L., R.S.; project administration, Q.X., T.J., T.L., W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Program of Science and Technology Department of Sichuan Province (No. 2022YFS0419 to T.L.); Sichuan Science and Technology Plan Projects—Key Research and Development Program (23ZDYF1772, awarded to T.J.; awarded to W.H.); National Natural Science Foundation of China (No. 82274321 to Q.X.; No. 81973632 to W.H.); National Institute for Health Research Senior Investigator Award, an EME Award (15/20/01), an MRC Award (MR/T002220/1) and the TransBioLine Consortium (all to R.S.).

Institutional Review Board Statement

The study did not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Iannuzzi, J.P.; King, J.A.; Leong, J.H.; Quan, J.; Windsor, J.W.; Tanyingoh, D.; Coward, S.; Forbes, N.; Heitman, S.J.; Shaheen, A.A.; et al. Global incidence of acute pancreatitis is increasing over time: A systematic review and meta-analysis. Gastroenterology 2022, 162, 122–134. [Google Scholar] [CrossRef]
  2. Li, C.L.; Jiang, M.; Pan, C.Q.; Li, J.; Xu, L.G. The global, regional, and national burden of acute pancreatitis in 204 countries and territories, 1990–2019. BMC Gastroenterol. 2021, 21, 332. [Google Scholar] [CrossRef] [PubMed]
  3. Szatmary, P.; Grammatikopoulos, T.; Cai, W.; Huang, W.; Mukherjee, R.; Halloran, C.; Beyer, G.; Sutton, R. Acute pancreatitis: Diagnosis and treatment. Drugs 2022, 82, 1251–1276. [Google Scholar] [CrossRef] [PubMed]
  4. Banks, P.A.; Bollen, T.L.; Dervenis, C.; Gooszen, H.G.; Johnson, C.D.; Sarr, M.G.; Tsiotos, G.G.; Vege, S.S.; Acute Pancreatitis Classification Working Group. Classification of acute pancreatitis--2012: Revision of the Atlanta classification and definitions by international consensus. Gut 2013, 62, 102–111. [Google Scholar] [CrossRef] [PubMed]
  5. Linkermann, A.; Stockwell, B.R.; Krautwald, S.; Anders, H.J. Regulated cell death and inflammation: An auto-amplification loop causes organ failure. Nat. Rev. Immunol. 2014, 14, 759–767. [Google Scholar] [CrossRef] [PubMed]
  6. Barreto, S.G.; Habtezion, A.; Gukovskaya, A.; Lugea, A.; Jeon, C.; Yadav, D.; Hegyi, P.; Venglovecz, V.; Sutton, R.; Pandol, S.J. Critical thresholds: Key to unlocking the door to the prevention and specific treatments for acute pancreatitis. Gut 2021, 70, 194–203. [Google Scholar] [CrossRef] [PubMed]
  7. Guo, Q.; Li, A.; Xia, Q.; Liu, X.; Tian, B.; Mai, G.; Huang, Z.; Chen, G.; Tang, W.; Jin, X.; et al. The role of organ failure and infection in necrotizing pancreatitis: A prospective study. Ann. Surg. 2014, 259, 1201–1207. [Google Scholar] [CrossRef] [PubMed]
  8. Sternby, H.; Bolado, F.; Canaval-Zuleta, H.J.; Marra-López, C.; Hernando-Alonso, A.I.; Del-Val-Antoñana, A.; García-Rayado, G.; Rivera-Irigoin, R.; Grau-García, F.J.; Oms, L.; et al. Determinants of severity in acute pancreatitis: A nation-wide multicenter prospective cohort study. Ann. Surg. 2019, 270, 348–355. [Google Scholar] [CrossRef] [PubMed]
  9. Schepers, N.J.; Bakker, O.J.; Besselink, M.G.; Ahmed Ali, U.; Bollen, T.L.; Gooszen, H.G.; van Santvoort, H.C.; Bruno, M.J. Impact of characteristics of organ failure and infected necrosis on mortality in necrotising pancreatitis. Gut 2019, 68, 1044–1051. [Google Scholar] [CrossRef]
  10. Shi, N.; Liu, T.; de la Iglesia-Garcia, D.; Deng, L.; Jin, T.; Lan, L.; Zhu, P.; Hu, W.; Zhou, Z.; Singh, V.; et al. Duration of organ failure impacts mortality in acute pancreatitis. Gut 2020, 69, 604–605. [Google Scholar] [CrossRef] [Green Version]
  11. Petrov, M.S.; Shanbhag, S.; Chakraborty, M.; Phillips, A.R.; Windsor, J.A. Organ failure and infection of pancreatic necrosis as determinants of mortality in patients with acute pancreatitis. Gastroenterology 2010, 139, 813–820. [Google Scholar] [CrossRef]
  12. Werge, M.; Novovic, S.; Schmidt, P.N.; Gluud, L.L. Infection increases mortality in necrotizing pancreatitis: A systematic review and meta-analysis. Pancreatology 2016, 16, 698–707. [Google Scholar] [CrossRef]
  13. Moggia, E.; Koti, R.; Belgaumkar, A.P.; Fazio, F.; Pereira, S.P.; Davidson, B.R.; Gurusamy, K.S. Pharmacological interventions for acute pancreatitis. Cochrane Database Syst. Rev. 2017, 4, Cd011384. [Google Scholar] [CrossRef] [PubMed]
  14. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 20 September 2022).
  15. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines; National Institutes of Health (US): Bethesda, MD, USA, 2021.
  16. Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Weber, A.; Barnes, B.J.; Egeblad, M.; et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, 5, e138999. [Google Scholar] [CrossRef]
  17. Carvalho, T.; Krammer, F.; Iwasaki, A. The first 12 months of COVID-19: A timeline of immunological insights. Nat. Rev. Immunol. 2021, 21, 245–256. [Google Scholar] [CrossRef] [PubMed]
  18. Chung, J.Y.; Thone, M.N.; Kwon, Y.J. COVID-19 vaccines: The status and perspectives in delivery points of view. Adv. Drug Deliv. Rev. 2021, 170, 1–25. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, F.; Long, X.; Zhang, B.; Zhang, W.; Chen, X.; Zhang, Z. ACE2 expression in pancreas may cause pancreatic damage after SARS-CoV-2 infection. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2020, 18, 2128–2130.e2. [Google Scholar] [CrossRef] [PubMed]
  20. Yang, F.; Xu, Y.; Dong, Y.; Huang, Y.; Fu, Y.; Li, T.; Sun, C.; Pandanaboyana, S.; Windsor, J.A.; Fu, D. Prevalence and prognosis of increased pancreatic enzymes in patients with COVID-19: A systematic review and meta-analysis. Pancreatology 2022, 22, 539–546. [Google Scholar] [CrossRef]
  21. Goyal, H.; Sachdeva, S.; Perisetti, A.; Mann, R.; Inamdar, S.; Tharian, B. Hyperlipasemia and potential pancreatic injury patterns in COVID-19: A marker of severity or innocent bystander? Gastroenterology 2021, 160, 946–948.e2. [Google Scholar] [CrossRef]
  22. Kiyak, M.; Düzenli, T. Lipase elevation on admission predicts worse clinical outcomes in patients with COVID-19. Pancreatology 2022, 22, 665–670. [Google Scholar] [CrossRef] [PubMed]
  23. Kumar, V.; Barkoudah, E.; Souza, D.A.T.; Jin, D.X.; McNabb-Baltar, J. Clinical course and outcome among patients with acute pancreatitis and COVID-19. Eur. J. Gastroenterol. Hepatol. 2021, 33, 695–700. [Google Scholar] [CrossRef]
  24. Szatmary, P.; Arora, A.; Thomas Raraty, M.G.; Joseph Dunne, D.F.; Baron, R.D.; Halloran, C.M. Emerging phenotype of severe acute respiratory syndrome-coronavirus 2-associated pancreatitis. Gastroenterology 2020, 159, 1551–1554. [Google Scholar] [CrossRef] [PubMed]
  25. De Bruyn, A.; Verellen, S.; Bruckers, L.; Geebelen, L.; Callebaut, I.; De Pauw, I.; Stessel, B.; Dubois, J. Secondary infection in COVID-19 critically ill patients: A retrospective single-center evaluation. BMC Infect. Dis. 2022, 22, 207. [Google Scholar] [CrossRef] [PubMed]
  26. Ripa, M.; Galli, L.; Poli, A.; Oltolini, C.; Spagnuolo, V.; Mastrangelo, A.; Muccini, C.; Monti, G.; De Luca, G.; Landoni, G.; et al. Secondary infections in patients hospitalized with COVID-19: Incidence and predictive factors. Clin. Microbiol. Infect. 2021, 27, 451–457. [Google Scholar] [CrossRef] [PubMed]
  27. Gallo, C.G.; Fiorino, S.; Posabella, G.; Antonacci, D.; Tropeano, A.; Pausini, E.; Pausini, C.; Guarniero, T.; Hong, W.; Giampieri, E.; et al. COVID-19, what could sepsis, severe acute pancreatitis, gender differences, and aging teach us? Cytokine 2021, 148, 155628. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, B.Z.; Sidell, M.A.; Wu, B.U.; Setiawan, V.W.; Chen, Z.; Xiang, A.H. Pre-existing pancreatitis and elevated risks of COVID-19 severity and mortality. Gastroenterology 2022, 162, 1758–1760.e3. [Google Scholar] [CrossRef]
  29. Karki, R.; Kanneganti, T.D. The ‘cytokine storm’: Molecular mechanisms and therapeutic prospects. Trends Immunol. 2021, 42, 681–705. [Google Scholar] [CrossRef]
  30. Guéant, J.L.; Guéant-Rodriguez, R.M.; Fromonot, J.; Oussalah, A.; Louis, H.; Chery, C.; Gette, M.; Gleye, S.; Callet, J.; Raso, J.; et al. Elastase and exacerbation of neutrophil innate immunity are involved in multi-visceral manifestations of COVID-19. Allergy 2021, 76, 1846–1858. [Google Scholar] [CrossRef]
  31. de Vries, F.; Huckriede, J.; Wichapong, K.; Reutelingsperger, C.; Nicolaes, G.A.F. The role of extracellular histones in COVID-19. J. Intern. Med. 2022, 293, 275–292. [Google Scholar] [CrossRef]
  32. Lee, P.J.; Papachristou, G.L. New insights into acute pancreatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 479–496. [Google Scholar] [CrossRef]
  33. Szatmary, P.; Huang, W.; Criddle, D.; Tepikin, A.; Sutton, R. Biology, role and therapeutic potential of circulating histones in acute inflammatory disorders. J. Cell Mol. Med. 2018, 22, 4617–4629. [Google Scholar] [CrossRef] [PubMed]
  34. Vora, S.M.; Lieberman, J.; Wu, H. Inflammasome activation at the crux of severe COVID-19. Nat. Rev. Immunol. 2021, 21, 694–703. [Google Scholar] [CrossRef] [PubMed]
  35. Naqvi, I.; Giroux, N.; Olson, L.; Morrison, S.A.; Llanga, T.; Akinade, T.O.; Zhu, Y.; Zhong, Y.; Bose, S.; Arvai, S.; et al. DAMPs/PAMPs induce monocytic TLR activation and tolerance in COVID-19 patients; nucleic acid binding scavengers can counteract such TLR agonists. Biomaterials 2022, 283, 121393. [Google Scholar] [CrossRef] [PubMed]
  36. Zheng, L.; Xue, J.; Jaffee, E.M.; Habtezion, A. Role of immune cells and immune-based therapies in pancreatitis and pancreatic ductal adenocarcinoma. Gastroenterology 2013, 144, 1230–1240. [Google Scholar] [CrossRef]
  37. Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S.; et al. Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell 2021, 184, 149–168.e17. [Google Scholar] [CrossRef] [PubMed]
  38. Li, X.; Zhang, Z.; Wang, Z.; Gutiérrez-Castrellón, P.; Shi, H. Cell deaths: Involvement in the pathogenesis and intervention therapy of COVID-19. Signal Transduct. Target. Ther. 2022, 7, 186. [Google Scholar] [CrossRef]
  39. Yamada, T.; Takaoka, A. Innate immune recognition against SARS-CoV-2. Inflamm. Regen. 2023, 43, 7. [Google Scholar] [CrossRef] [PubMed]
  40. Habtezion, A.; Gukovskaya, A.S.; Pandol, S.J. Acute pancreatitis: A multifaceted set of organelle and cellular interactions. Gastroenterology 2019, 156, 1941–1950. [Google Scholar] [CrossRef]
  41. Garg, P.K.; Singh, V.P. Organ failure due to systemic injury in acute pancreatitis. Gastroenterology 2019, 156, 2008–2023. [Google Scholar] [CrossRef]
  42. Xue, J.; Sharma, V.; Habtezion, A. Immune cells and immune-based therapy in pancreatitis. Immunol. Res. 2014, 58, 378–386. [Google Scholar] [CrossRef]
  43. Liu, S.; Szatmary, P.; Lin, J.W.; Wang, Q.; Sutton, R.; Chen, L.; Liu, T.; Huang, W.; Xia, Q. Circulating monocytes in acute pancreatitis. Front. Immunol. 2022, 13, 1062849. [Google Scholar] [CrossRef]
  44. Felsenstein, S.; Herbert, J.A.; McNamara, P.S.; Hedrich, C.M. COVID-19: Immunology and treatment options. Clin. Immunol. 2020, 215, 108448. [Google Scholar] [CrossRef] [PubMed]
  45. Fonteh, P.; Smith, M.; Brand, M. Adaptive immune cell dysregulation and role in acute pancreatitis disease progression and treatment. Arch. Immunol. Et Ther. Exp. 2018, 66, 199–209. [Google Scholar] [CrossRef] [PubMed]
  46. Li, G.; Fan, Y.; Lai, Y.; Han, T.; Li, Z.; Zhou, P.; Pan, P.; Wang, W.; Hu, D.; Liu, X.; et al. Coronavirus infections and immune responses. J. Med. Virol. 2020, 92, 424–432. [Google Scholar] [CrossRef] [PubMed]
  47. Anka, A.U.; Tahir, M.I.; Abubakar, S.D.; Alsabbagh, M.; Zian, Z.; Hamedifar, H.; Sabzevari, A.; Azizi, G. Coronavirus disease 2019 (COVID-19): An overview of the immunopathology, serological diagnosis and management. Scand. J. Immunol. 2021, 93, e12998. [Google Scholar] [CrossRef]
  48. Pietruczuk, M.; Dabrowska, M.I.; Wereszczynska-Siemiatkowska, U.; Dabrowski, A. Alteration of peripheral blood lymphocyte subsets in acute pancreatitis. World J. Gastroenterol. 2006, 12, 5344–5351. [Google Scholar] [CrossRef] [PubMed]
  49. Norman, J. The role of cytokines in the pathogenesis of acute pancreatitis. Am. J. Surg. 1998, 175, 76–83. [Google Scholar] [CrossRef]
  50. Makhija, R.; Kingsnorth, A.N. Cytokine storm in acute pancreatitis. J. Hepato-Biliary-Pancreat. Surg. 2002, 9, 401–410. [Google Scholar] [CrossRef]
  51. To, K.K.; Sridhar, S.; Chiu, K.H.; Hung, D.L.; Li, X.; Hung, I.F.; Tam, A.R.; Chung, T.W.; Chan, J.F.; Zhang, A.J.; et al. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg. Microbes Infect. 2021, 10, 507–535. [Google Scholar] [CrossRef]
  52. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469. [Google Scholar] [CrossRef]
  53. Hegyi, P.; Szakács, Z.; Sahin-Tóth, M. Lipotoxicity and cytokine storm in severe acute pancreatitis and COVID-19. Gastroenterology 2020, 159, 824–827. [Google Scholar] [CrossRef] [PubMed]
  54. Pena, A.L.B.; Oliveira, R.A.; Severo, R.G.; Simões, E.S.A.C. COVID-19 related coagulopathy: What is Known Up to Now. Curr. Med. Chem. 2021, 28, 4207–4225. [Google Scholar] [CrossRef] [PubMed]
  55. Subramaniam, S.; Kothari, H.; Bosmann, M. Tissue factor in COVID-19-associated coagulopathy. Thromb. Res. 2022, 220, 35–47. [Google Scholar] [CrossRef]
  56. Bettac, L.; Denk, S.; Seufferlein, T.; Huber-Lang, M. Complement in pancreatic disease-perpetrator or savior? Front. Immunol. 2017, 8, 15. [Google Scholar] [CrossRef] [PubMed]
  57. Dumnicka, P.; Maduzia, D.; Ceranowicz, P.; Olszanecki, R.; Drożdż, R.; Kuśnierz-Cabala, B. The interplay between inflammation, coagulation and endothelial injury in the early phase of acute pancreatitis: Clinical implications. Int. J. Mol. Sci. 2017, 18, 354. [Google Scholar] [CrossRef]
  58. Kakafika, A.; Papadopoulos, V.; Mimidis, K.; Mikhailidis, D.P. Coagulation, platelets, and acute pancreatitis. Pancreas 2007, 34, 15–20. [Google Scholar] [CrossRef] [PubMed]
  59. El-Kurdi, B.; Khatua, B.; Rood, C.; Snozek, C.; Cartin-Ceba, R.; Singh, V.P. Mortality from coronavirus disease 2019 increases with unsaturated fat and may be reduced by early calcium and albumin supplementation. Gastroenterology 2020, 159, 1015–1018.e4. [Google Scholar] [CrossRef] [PubMed]
  60. Saccon, T.D.; Mousovich-Neto, F.; Ludwig, R.G.; Carregari, V.C.; Dos Anjos Souza, A.B.; Dos Passos, A.S.C.; Martini, M.C.; Barbosa, P.P.; de Souza, G.F.; Muraro, S.P.; et al. SARS-CoV-2 infects adipose tissue in a fat depot- and viral lineage-dependent manner. Nat. Commun. 2022, 13, 5722. [Google Scholar] [CrossRef]
  61. Patel, K.; Trivedi, R.N.; Durgampudi, C.; Noel, P.; Cline, R.A.; DeLany, J.P.; Navina, S.; Singh, V.P. Lipolysis of visceral adipocyte triglyceride by pancreatic lipases converts mild acute pancreatitis to severe pancreatitis independent of necrosis and inflammation. Am. J. Pathol. 2015, 185, 808–819. [Google Scholar] [CrossRef] [PubMed]
  62. de Oliveira, C.; Khatua, B.; Noel, P.; Kostenko, S.; Bag, A.; Balakrishnan, B.; Patel, K.S.; Guerra, A.A.; Martinez, M.N.; Trivedi, S.; et al. Pancreatic triglyceride lipase mediates lipotoxic systemic inflammation. J. Clin. Investig. 2020, 130, 1931–1947. [Google Scholar] [CrossRef] [Green Version]
  63. Navina, S.; Acharya, C.; DeLany, J.P.; Orlichenko, L.S.; Baty, C.J.; Shiva, S.S.; Durgampudi, C.; Karlsson, J.M.; Lee, K.; Bae, K.T.; et al. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci. Transl. Med. 2011, 3, 107ra110. [Google Scholar] [CrossRef] [PubMed]
  64. Baek, Y.B.; Kwon, H.J.; Sharif, M.; Lim, J.; Lee, I.C.; Ryu, Y.B.; Lee, J.I.; Kim, J.S.; Lee, Y.S.; Kim, D.H.; et al. Therapeutic strategy targeting host lipolysis limits infection by SARS-CoV-2 and influenza A virus. Signal Transduct. Target. Ther. 2022, 7, 367. [Google Scholar] [CrossRef] [PubMed]
  65. Meakins, J.L.; Pietsch, J.B.; Bubenick, O.; Kelly, R.; Rode, H.; Gordon, J.; MacLean, L.D. Delayed hypersensitivity: Indicator of acquired failure of host defenses in sepsis and trauma. Ann. Surg. 1977, 186, 241–250. [Google Scholar] [CrossRef]
  66. MacLean, L.D.; Meakins, J.L.; Taguchi, K.; Duignan, J.P.; Dhillon, K.S.; Gordon, J. Host resistance in sepsis and trauma. Ann. Surg. 1975, 182, 207–217. [Google Scholar] [CrossRef]
  67. Monneret, G.; Gossez, M.; Aghaeepour, N.; Gaudilliere, B.; Venet, F. How clinical flow cytometry rebooted sepsis immunology. Cytom. Part A J. Int. Soc. Anal. Cytol. 2019, 95, 431–441. [Google Scholar] [CrossRef] [PubMed]
  68. Garcia-Sabrido, J.L.; Valdecantos, E.; Bastida, E.; Tellado, J.M. The anergic state as a predictor of pancreatic sepsis. Zent. Fur Chir. 1989, 114, 114–120. [Google Scholar]
  69. Mayerle, J.; Dummer, A.; Sendler, M.; Malla, S.R.; van den Brandt, C.; Teller, S.; Aghdassi, A.; Nitsche, C.; Lerch, M.M. Differential roles of inflammatory cells in pancreatitis. J. Gastroenterol. Hepatol. 2012, 27 (Suppl. 2), 47–51. [Google Scholar] [CrossRef]
  70. Jain, S.; Midha, S.; Mahapatra, S.J.; Gupta, S.; Sharma, M.K.; Nayak, B.; Jacob, T.G.; Shalimar; Garg, P.K. Interleukin-6 significantly improves predictive value of systemic inflammatory response syndrome for predicting severe acute pancreatitis. Pancreatology 2018, 18, 500–506. [Google Scholar] [CrossRef] [PubMed]
  71. Bhatia, M. Acute pancreatitis as a model of SIRS. Front. Biosci. (Landmark Ed.) 2009, 14, 2042–2050. [Google Scholar] [CrossRef]
  72. Bone, R.C. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit. Care Med. 1996, 24, 1125–1128. [Google Scholar] [CrossRef]
  73. Zhulai, G.A.; Oleinik, E.K.; Ostrovskii, K.A.; Oleinik, V.M.; Kravchenko, P.N.; Churov, A.V. Alterations of lymphocyte subsets and indicators of immune suppression in patients with acute pancreatitis. Eksp. Klin. Gastroenterol. 2014, 9, 21–25. [Google Scholar]
  74. Döcke, W.D.; Randow, F.; Syrbe, U.; Krausch, D.; Asadullah, K.; Reinke, P.; Volk, H.D.; Kox, W. Monocyte deactivation in septic patients: Restoration by IFN-gamma treatment. Nat. Med. 1997, 3, 678–681. [Google Scholar] [CrossRef]
  75. Volk, H.D.; Reinke, P.; Krausch, D.; Zuckermann, H.; Asadullah, K.; Müller, J.M.; Döcke, W.D.; Kox, W.J. Monocyte deactivation--rationale for a new therapeutic strategy in sepsis. Intensive Care Med. 1996, 22 (Suppl. 4), S474–S481. [Google Scholar] [CrossRef] [PubMed]
  76. Venet, F.; Lepape, A.; Monneret, G. Clinical review: Flow cytometry perspectives in the ICU-from diagnosis of infection to monitoring of injury-induced immune dysfunctions. Crit. Care 2011, 15, 231–239. [Google Scholar] [CrossRef] [PubMed]
  77. Hotchkiss, R.S.; Monneret, G.; Payen, D. Immunosuppression in sepsis: A novel understanding of the disorder and a new therapeutic approach. Lancet Infect. Dis. 2013, 13, 260–268. [Google Scholar] [CrossRef] [PubMed]
  78. Leentjens, J.; Kox, M.; van der Hoeven, J.G.; Netea, M.G.; Pickkers, P. Immunotherapy for the adjunctive treatment of sepsis: From immunosuppression to immunostimulation. Time for a paradigm change? Am. J. Respir. Crit. Care Med. 2013, 187, 1287–1293. [Google Scholar] [CrossRef] [PubMed]
  79. Giamarellos-Bourboulis, E.J. What is the pathophysiology of the septic host upon admission? Int. J. Antimicrob. Agents 2010, 36 (Suppl. 2), S2–S5. [Google Scholar] [CrossRef] [PubMed]
  80. Minkov, G.A.; Halacheva, K.S.; Yovtchev, Y.P.; Gulubova, M.V. Pathophysiological mechanisms of acute pancreatitis define inflammatory markers of clinical prognosis. Pancreas 2015, 44, 713–717. [Google Scholar] [CrossRef]
  81. Zhuang, Y.G.; Peng, H.; Chen, Y.Z.; Zhou, S.Q.; Chen, Y.Q. Dynamic monitoring of monocyte HLA-DR expression for the diagnosis, prognosis, and prediction of sepsis. Front. Biosci.-Landmrk 2017, 22, 1344–1354. [Google Scholar]
  82. Li, J.; Yang, W.-J.; Huang, L.-M.; Tang, C.-W. Immunomodulatory therapies for acute pancreatitis. World J. Gastroenterol. 2014, 20, 16935–16947. [Google Scholar] [CrossRef] [PubMed]
  83. Misra, A.K.; Levy, M.M.; Ward, N.S. Biomarkers of Immunosuppression. Crit. Care Clin. 2020, 36, 167–176. [Google Scholar] [CrossRef]
  84. Benlyamani, I.; Venet, F.; Coudereau, R.; Gossez, M.; Monneret, G. Monocyte HLA-DR measurement by flow cytometry in COVID-19 patients: An interim review. Cytom. Part A J. Int. Soc. Anal. Cytol. 2020, 97, 1217–1221. [Google Scholar] [CrossRef] [PubMed]
  85. Manohar, M.; Jones, E.K.; Rubin, S.J.S.; Subrahmanyam, P.B.; Swaminathan, G.; Mikhail, D.; Bai, L.; Singh, G.; Wei, Y.; Sharma, V.; et al. Novel circulating and tissue monocytes as well as macrophages in pancreatitis and recovery. Gastroenterology 2021, 161, 2014–2029.e14. [Google Scholar] [CrossRef] [PubMed]
  86. Napoli, C.; Benincasa, G.; Criscuolo, C.; Faenza, M.; Liberato, C.; Rusciano, M. Immune reactivity during COVID-19: Implications for treatment. Immunol. Lett. 2021, 231, 28–34. [Google Scholar] [CrossRef] [PubMed]
  87. Volk, H.D.; Thieme, M.; Heym, S.; Döcke, W.D.; Ruppe, U.; Tausch, W.; Manger, D.; Zuckermann, S.; Golosubow, A.; Nieter, B.; et al. Alterations in function and phenotype of monocytes from patients with septic disease--predictive value and new therapeutic strategies. Behring. Inst. Mitt. 1991, 88, 208–215. [Google Scholar]
  88. Lin, R.Y.; Astiz, M.E.; Saxon, J.C.; Rackow, E.C. Altered leukocyte immunophenotypes in septic shock. Studies of HLA-DR, CD11b, CD14, and IL-2R expression. Chest 1993, 104, 847–853. [Google Scholar] [CrossRef] [PubMed]
  89. Ditschkowski, M.; Kreuzfelder, E.; Rebmann, V.; Ferencik, S.; Majetschak, M.; Schmid, E.N.; Obertacke, U.; Hirche, H.; Schade, U.F.; Grosse-Wilde, H. HLA-DR expression and soluble HLA-DR levels in septic patients after trauma. Ann. Surg. 1999, 229, 246–254. [Google Scholar] [CrossRef]
  90. Wakefield, C.H.; Carey, P.D.; Foulds, S.; Monson, J.R.; Guillou, P.J. Changes in major histocompatibility complex class II expression in monocytes and T cells of patients developing infection after surgery. Br. J. Surg. 1993, 80, 205–209. [Google Scholar] [CrossRef] [PubMed]
  91. Sachse, C.; Prigge, M.; Cramer, G.; Pallua, N.; Henkel, E. Association between reduced human leukocyte antigen (HLA)-DR expression on blood monocytes and increased plasma level of interleukin-10 in patients with severe burns. Clin. Chem. Lab. Med. 1999, 37, 193–198. [Google Scholar] [CrossRef] [PubMed]
  92. Kaufman, J.F.; Auffray, C.; Korman, A.J.; Shackelford, D.A.; Strominger, J. The class II molecules of the human and murine major histocompatibility complex. Cell 1984, 36, 1–13. [Google Scholar] [CrossRef]
  93. Unanue, E.R.; Turk, V.; Neefjes, J. Variations in MHC Class II antigen processing and presentation in health and disease. Annu. Rev. Immunol. 2016, 34, 265–297. [Google Scholar] [CrossRef]
  94. Crux, N.B.; Elahi, S. Human leukocyte antigen (HLA) and immune regulation: How do classical and non-classical HLA alleles modulate immune response to human immunodeficiency virus and hepatitis C virus infections? Front. Immunol. 2017, 8, 832. [Google Scholar] [CrossRef]
  95. Mosaad, Y.M. Clinical role of human leukocyte antigen in health and disease. Scand. J. Immunol. 2015, 82, 283–306. [Google Scholar] [CrossRef]
  96. Monneret, G.; Venet, F. Sepsis-induced immune alterations monitoring by flow cytometry as a promising tool for individualized therapy. Cytom. B Clin. Cytom. 2016, 90, 376–386. [Google Scholar] [CrossRef]
  97. Monneret, G.; Venet, F. Monocyte HLA-DR in sepsis: Shall we stop following the flow? Crit. Care 2014, 18, 102. [Google Scholar] [CrossRef]
  98. Monneret, G.; Elmenkouri, N.; Bohe, J.; Debard, A.L.; Gutowski, M.C.; Bienvenu, J.; Lepape, A. Analytical requirements for measuring monocytic human lymphocyte antigen DR by flow cytometry: Application to the monitoring of patients with septic shock. Clin. Chem. 2002, 48, 1589–1592. [Google Scholar] [CrossRef] [PubMed]
  99. Docke, W.D.; Hoflich, C.; Davis, K.A.; Rottgers, K.; Meisel, C.; Kiefer, P.; Weber, S.U.; Hedwig-Geissing, M.; Kreuzfelder, E.; Tschentscher, P.; et al. Monitoring temporary immunodepression by flow cytometric measurement of monocytic HLA-DR expression: A multicenter standardized study. Clin. Chem. 2005, 51, 2341–2347. [Google Scholar] [CrossRef] [PubMed]
  100. Demaret, J.; Walencik, A.; Jacob, M.C.; Timsit, J.F.; Venet, F.; Lepape, A.; Monneret, G. Inter-laboratory assessment of flow cytometric monocyte HLA-DR expression in clinical samples. Cytom. B Clin. Cytom. 2013, 84, 59–62. [Google Scholar] [CrossRef] [PubMed]
  101. Cajander, S.; Backman, A.; Tina, E.; Stralin, K.; Soderquist, B.; Kallman, J. Preliminary results in quantitation of HLA-DRA by real-time PCR: A promising approach to identify immunosuppression in sepsis. Crit. Care 2013, 17, R223. [Google Scholar] [CrossRef] [PubMed]
  102. van den Elsen, P.J. Expression regulation of major histocompatibility complex class I and class II encoding genes. Front. Immunol. 2011, 2, 48. [Google Scholar] [CrossRef]
  103. Steimle, V.; Otten, L.A.; Zufferey, M.; Mach, B. Complementation cloning of an MHC class II transactivator mutated in hereditary MHC class II deficiency (or bare lymphocyte syndrome). Cell 1993, 75, 135–146. [Google Scholar] [CrossRef] [PubMed]
  104. Miatello, J.; Lukaszewicz, A.C.; Carter, M.J.; Faivre, V.; Hua, S.; Martinet, K.Z.; Bourgeois, C.; Quintana-Murci, L.; Payen, D.; Boniotto, M.; et al. CIITA promoter polymorphism impairs monocytes HLA-DR expression in patients with septic shock. iScience 2022, 25, 105291. [Google Scholar] [CrossRef] [PubMed]
  105. Pishesha, N.; Harmand, T.J.; Ploegh, H.L. A guide to antigen processing and presentation. Nat. Rev. Immunol. 2022, 22, 751–764. [Google Scholar] [CrossRef] [PubMed]
  106. Viville, S.; Neefjes, J.; Lotteau, V.; Dierich, A.; Lemeur, M.; Ploegh, H.; Benoist, C.; Mathis, D. Mice lacking the MHC class II-associated invariant chain. Cell 1993, 72, 635–648. [Google Scholar] [CrossRef] [PubMed]
  107. Wolk, K.; Kunz, S.; Crompton, N.E.; Volk, H.D.; Sabat, R. Multiple mechanisms of reduced major histocompatibility complex class II expression in endotoxin tolerance. J. Biol. Chem. 2003, 278, 18030–18036. [Google Scholar] [CrossRef] [PubMed]
  108. Alfonso, C.; Karlsson, L. Nonclassical MHC class II molecules. Annu. Rev. Immunol. 2000, 18, 113–142. [Google Scholar] [CrossRef] [PubMed]
  109. Fung-Leung, W.P.; Surh, C.D.; Liljedahl, M.; Pang, J.; Leturcq, D.; Peterson, P.A.; Webb, S.R.; Karlsson, L. Antigen presentation and T cell development in H2-M-deficient mice. Science 1996, 271, 1278–1281. [Google Scholar] [CrossRef]
  110. Pinet, V.; Vergelli, M.; Martin, R.; Bakke, O.; Long, E.O. Antigen presentation mediated by recycling of surface HLA-DR molecules. Nature 1995, 375, 603–606. [Google Scholar] [CrossRef]
  111. Neefjes, J.; Jongsma, M.L.M.; Paul, P.; Bakke, O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat. Rev. Immunol. 2011, 11, 823–836. [Google Scholar] [CrossRef]
  112. Koppelman, B.; Neefjes, J.J.; de Vries, J.E.; de Waal Malefyt, R. Interleukin-10 down-regulates MHC class II alphabeta peptide complexes at the plasma membrane of monocytes by affecting arrival and recycling. Immunity 1997, 7, 861–871. [Google Scholar] [CrossRef]
  113. Ting, J.P.; Trowsdale, J. Genetic control of MHC class II expression. Cell 2002, 109 (Suppl. 1), S21–S33. [Google Scholar] [CrossRef]
  114. Czarniecki, C.W.; Chiu, H.H.; Wong, G.H.; McCabe, S.M.; Palladino, M.A. Transforming growth factor-beta 1 modulates the expression of class II histocompatibility antigens on human cells. J. Immunol. 1988, 140, 4217–4223. [Google Scholar] [CrossRef]
  115. Piskurich, J.F.; Wang, Y.; Linhoff, M.W.; White, L.C.; Ting, J.P. Identification of distinct regions of 5’ flanking DNA that mediate constitutive, IFN-gamma, STAT1, and TGF-beta-regulated expression of the class II transactivator gene. J. Immunol. 1998, 160, 233–240. [Google Scholar] [CrossRef]
  116. Lu, H.T.; Riley, J.L.; Babcock, G.T.; Huston, M.; Stark, G.R.; Boss, J.M.; Ransohoff, R.M. Interferon (IFN) beta acts downstream of IFN-gamma-induced class II transactivator messenger RNA accumulation to block major histocompatibility complex class II gene expression and requires the 48-kD DNA-binding protein, ISGF3-gamma. J. Exp. Med. 1995, 182, 1517–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hershman, M.J.; Appel, S.H.; Wellhausen, S.R.; Sonnenfeld, G.; Polk, H.C., Jr. Interferon-gamma treatment increases HLA-DR expression on monocytes in severely injured patients. Clin Exp Immunol 1989, 77, 67–70. [Google Scholar] [PubMed]
  118. Blanar, M.A.; Boettger, E.C.; Flavell, R.A. Transcriptional activation of HLA-DR alpha by interferon gamma requires a trans-acting protein. Proc. Natl. Acad. Sci. USA 1988, 85, 4672–4676. [Google Scholar] [CrossRef] [PubMed]
  119. Pierre, P.; Turley, S.J.; Gatti, E.; Hull, M.; Meltzer, J.; Mirza, A.; Inaba, K.; Steinman, R.M.; Mellman, I. Developmental regulation of MHC class II transport in mouse dendritic cells. Nature 1997, 388, 787–792. [Google Scholar] [CrossRef] [PubMed]
  120. Cella, M.; Engering, A.; Pinet, V.; Pieters, J.; Lanzavecchia, A. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells. Nature 1997, 388, 782–787. [Google Scholar] [CrossRef] [PubMed]
  121. Hotchkiss, R.S.; Monneret, G.; Payen, D. Sepsis-induced immunosuppression: From cellular dysfunctions to immunotherapy. Nat. Rev. Immunol. 2013, 13, 862–874. [Google Scholar] [CrossRef] [PubMed]
  122. Cao, C.; Yu, M.; Chai, Y. Pathological alteration and therapeutic implications of sepsis-induced immune cell apoptosis. Cell Death Dis. 2019, 10, 782. [Google Scholar] [CrossRef] [PubMed]
  123. Kylanpaa-Back, M.L.; Takala, A.; Kemppainen, E.; Puolakkainen, P.; Kautiainen, H.; Jansson, S.E.; Haapiainen, R.; Repo, H. Cellular markers of systemic inflammation and immune suppression in patients with organ failure due to severe acute pancreatitis. Scand. J. Gastroenterol. 2001, 36, 1100–1107. [Google Scholar]
  124. Richter, A.; Nebe, T.; Wendl, K.; Schuster, K.; Klaebisch, G.; Quintel, M.; Lorenz, D.; Post, S.; Trede, M. HLA-DR expression in acute pancreatitis. Eur. J. Surg. 1999, 165, 947–951. [Google Scholar] [CrossRef] [PubMed]
  125. Turunen, A.; Kuuliala, A.; Penttilä, A.; Kaukonen, K.M.; Mustonen, H.; Pettilä, V.; Puolakkainen, P.; Kylänpää, L.; Kuuliala, K. Time course of signaling profiles of blood leukocytes in acute pancreatitis and sepsis. Scand. J. Clin. Lab. Investig. 2020, 80, 114–123. [Google Scholar] [CrossRef] [PubMed]
  126. Li, J.P.; Yang, J.; Huang, J.R.; Jiang, D.L.; Zhang, F.; Liu, M.F.; Qiang, Y.; Gu, Y.L. Immunosuppression and the infection caused by gut mucosal barrier dysfunction in patients with early severe acute pancreatitis. Front. Biosci. (Landmark Ed.) 2013, 18, 892–900. [Google Scholar] [PubMed]
  127. Mentula, P.; Kylanpaa, M.L.; Kemppainen, E.; Jansson, S.E.; Sarna, S.; Puolakkainen, P.; Haapiainen, R.; Repo, H. Plasma anti-inflammatory cytokines and monocyte human leucocyte antigen-DR expression in patients with acute pancreatitis. Scand. J. Gastroenterol. 2004, 39, 178–187. [Google Scholar] [CrossRef] [PubMed]
  128. Yu, W.K.; Li, W.Q.; Li, N.; Li, J.S. Mononuclear histocompatibility leukocyte antigen-DR expression in the early phase of acute pancreatitis. Pancreatology 2004, 4, 233–243. [Google Scholar] [CrossRef]
  129. Walter, L.O.; Cardoso, C.C.; Santos-Pirath, Í.M.; Costa, H.Z.; Gartner, R.; Werle, I.; Mohr, E.T.B.; da Rosa, J.S.; Felisberto, M.; Kretzer, I.F.; et al. The relationship between peripheral immune response and disease severity in SARS-CoV-2-infected subjects: A cross-sectional study. Immunology 2022, 165, 481–496. [Google Scholar] [CrossRef] [PubMed]
  130. Yang, Q.; Wen, Y.; Qi, F.; Gao, X.; Chen, W.; Xu, G.; Wei, C.; Wang, H.; Tang, X.; Lin, J.; et al. Suppressive monocytes impair MAIT cells response via IL-10 in patients with severe COVID-19. J. Immunol. 2021, 207, 1848–1856. [Google Scholar] [CrossRef] [PubMed]
  131. Gatti, A.; Radrizzani, D.; Viganò, P.; Mazzone, A.; Brando, B. Decrease of non-classical and intermediate monocyte subsets in severe acute SARS-CoV-2 infection. Cytom. Part A J. Int. Soc. Anal. Cytol. 2020, 97, 887–890. [Google Scholar] [CrossRef]
  132. Mudd, P.A.; Crawford, J.C.; Turner, J.S.; Souquette, A.; Reynolds, D.; Bender, D.; Bosanquet, J.P.; Anand, N.J.; Striker, D.A.; Martin, R.S.; et al. Distinct inflammatory profiles distinguish COVID-19 from influenza with limited contributions from cytokine storm. Sci. Adv. 2020, 6, eabe3024. [Google Scholar] [CrossRef]
  133. Xu, G.; Qi, F.; Li, H.; Yang, Q.; Wang, H.; Wang, X.; Liu, X.; Zhao, J.; Liao, X.; Liu, Y.; et al. The differential immune responses to COVID-19 in peripheral and lung revealed by single-cell RNA sequencing. Cell Discov. 2020, 6, 73. [Google Scholar] [CrossRef]
  134. Bonnet, B.; Cosme, J.; Dupuis, C.; Coupez, E.; Adda, M.; Calvet, L.; Fabre, L.; Saint-Sardos, P.; Bereiziat, M.; Vidal, M.; et al. Severe COVID-19 is characterized by the co-occurrence of moderate cytokine inflammation and severe monocyte dysregulation. EBioMedicine 2021, 73, 103622. [Google Scholar] [CrossRef] [PubMed]
  135. Christensen, E.E.; Jørgensen, M.J.; Nore, K.G.; Dahl, T.B.; Yang, K.; Ranheim, T.; Huse, C.; Lind, A.; Nur, S.; Stiksrud, B.; et al. Critical COVID-19 is associated with distinct leukocyte phenotypes and transcriptome patterns. J. Intern. Med. 2021, 290, 677–692. [Google Scholar] [CrossRef] [PubMed]
  136. Qin, S.; Jiang, Y.; Wei, X.; Liu, X.; Guan, J.; Chen, Y.; Lu, H.; Qian, J.; Wang, Z.; Lin, X. Dynamic changes in monocytes subsets in COVID-19 patients. Hum. Immunol. 2021, 82, 170–176. [Google Scholar] [CrossRef] [PubMed]
  137. Zenarruzabeitia, O.; Astarloa-Pando, G.; Terrén, I.; Orrantia, A.; Pérez-Garay, R.; Seijas-Betolaza, I.; Nieto-Arana, J.; Imaz-Ayo, N.; Pérez-Fernández, S.; Arana-Arri, E.; et al. T cell activation, highly armed cytotoxic cells and a shift in monocytes CD300 receptors expression is characteristic of patients with severe COVID-19. Front. Immunol. 2021, 12, 655934. [Google Scholar] [CrossRef] [PubMed]
  138. Cizmecioglu, A.; Emsen, A.; Sumer, S.; Ergun, D.; Akay Cizmecioglu, H.; Turk Dagi, H.; Artac, H. Reduced monocyte subsets, their HLA-DR expressions, and relations to acute phase reactants in severe COVID-19 cases. Viral Immunol. 2022, 35, 273–282. [Google Scholar] [CrossRef] [PubMed]
  139. Hammad, R.; Kotb, H.G.; Eldesoky, G.A.; Mosaad, A.M.; El-Nasser, A.M.; Abd El Hakam, F.E.; Eldesoky, N.A.; Mashaal, A.; Farhoud, H. Utility of monocyte expression of HLA-DR versus T lymphocyte frequency in the assessment of COVID-19 outcome. Int. J. Gen. Med. 2022, 15, 5073–5087. [Google Scholar] [CrossRef] [PubMed]
  140. Venet, F.; Gossez, M.; Bidar, F.; Bodinier, M.; Coudereau, R.; Lukaszewicz, A.C.; Tardiveau, C.; Brengel-Pesce, K.; Cheynet, V.; Cazalis, M.A.; et al. T cell response against SARS-CoV-2 persists after one year in patients surviving severe COVID-19. EBioMedicine 2022, 78, 103967. [Google Scholar] [CrossRef]
  141. Marais, C.; Claude, C.; Semaan, N.; Charbel, R.; Barreault, S.; Travert, B.; Piloquet, J.E.; Demailly, Z.; Morin, L.; Merchaoui, Z.; et al. Myeloid phenotypes in severe COVID-19 predict secondary infection and mortality: A pilot study. Ann. Intensive Care 2021, 11, 111. [Google Scholar] [CrossRef]
  142. Giamarellos-Bourboulis, E.J.; Netea, M.G.; Rovina, N.; Akinosoglou, K.; Antoniadou, A.; Antonakos, N.; Damoraki, G.; Gkavogianni, T.; Adami, M.E.; Katsaounou, P.; et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe 2020, 27, 992–1000.e3. [Google Scholar] [CrossRef]
  143. Moser, D.; Biere, K.; Han, B.; Hoerl, M.; Schelling, G.; Choukér, A.; Woehrle, T. COVID-19 impairs immune response to candida albicans. Front. Immunol. 2021, 12, 640644. [Google Scholar] [CrossRef]
  144. Utrero-Rico, A.; González-Cuadrado, C.; Chivite-Lacaba, M.; Cabrera-Marante, O.; Laguna-Goya, R.; Almendro-Vazquez, P.; Díaz-Pedroche, C.; Ruiz-Ruigómez, M.; Lalueza, A.; Folgueira, M.D.; et al. Alterations in circulating monocytes predict COVID-19 severity and include chromatin modifications still detectable six months after recovery. Biomedicines 2021, 9, 1253. [Google Scholar] [CrossRef] [PubMed]
  145. Mairpady Shambat, S.; Gómez-Mejia, A.; Schweizer, T.A.; Huemer, M.; Chang, C.C.; Acevedo, C.; Bergada-Pijuan, J.; Vulin, C.; Hofmaenner, D.A.; Scheier, T.C.; et al. Hyperinflammatory environment drives dysfunctional myeloid cell effector response to bacterial challenge in COVID-19. PLoS Pathog. 2022, 18, e1010176. [Google Scholar] [CrossRef] [PubMed]
  146. Boumaza, A.; Gay, L.; Mezouar, S.; Bestion, E.; Diallo, A.B.; Michel, M.; Desnues, B.; Raoult, D.; La Scola, B.; Halfon, P.; et al. Monocytes and macrophages, targets of severe acute respiratory syndrome coronavirus 2: The clue for coronavirus disease 2019 immunoparalysis. J. Infect. Dis. 2021, 224, 395–406. [Google Scholar] [CrossRef]
  147. Laing, A.G.; Lorenc, A.; Del Molino Del Barrio, I.; Das, A.; Fish, M.; Monin, L.; Muñoz-Ruiz, M.; McKenzie, D.R.; Hayday, T.S.; Francos-Quijorna, I.; et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef] [PubMed]
  148. Penttilä, P.A.; Van Gassen, S.; Panovska, D.; Vanderbeke, L.; Van Herck, Y.; Quintelier, K.; Emmaneel, A.; Filtjens, J.; Malengier-Devlies, B.; Ahmadzadeh, K.; et al. High dimensional profiling identifies specific immune types along the recovery trajectories of critically ill COVID19 patients. Cell Mol. Life Sci. 2021, 78, 3987–4002. [Google Scholar] [CrossRef]
  149. Zhang, R.; Shi, J.; Zhang, R.; Ni, J.; Habtezion, A.; Wang, X.; Hu, G.; Xue, J. Expanded CD14(hi)CD16(-) Immunosuppressive monocytes predict disease severity in patients with acute pancreatitis. J. Immunol. 2019, 202, 2578–2584. [Google Scholar] [CrossRef]
  150. Minkov, G.; Dimitrov, E.; Yovtchev, Y.; Enchev, E.; Lokova, R.; Halacheva, K. Prognostic value of peripheral blood CD14+HLA-DR+ monocytes in patients with acute pancreatitis. J. Immunoass. Immunochem. 2021, 42, 478–492. [Google Scholar] [CrossRef] [PubMed]
  151. Mentula, P.; Kylanpaa-Back, M.L.; Kemppainen, E.; Takala, A.; Jansson, S.E.; Kautiainen, H.; Puolakkainen, P.; Haapiainen, R.; Repo, H. Decreased HLA (human leucocyte antigen)-DR expression on peripheral blood monocytes predicts the development of organ failure in patients with acute pancreatitis. Clin. Sci. 2003, 105, 409–417. [Google Scholar] [CrossRef]
  152. Ho, Y.P.; Sheen, I.S.; Chiu, C.T.; Wu, C.S.; Lin, C.Y. A strong association between down-regulation of HLA-DR expression and the late mortality in patients with severe acute pancreatitis. Am. J. Gastroenterol. 2006, 101, 1117–1124. [Google Scholar] [CrossRef]
  153. Yang, M.; Liu, Z.; Kang, K.; Yu, K.; Wang, C. Letter to the Editor: CD14(+)HLA-DR(+) Cells in patients may be a biomarker reflecting the progression of COVID-19. Viral Immunol. 2021, 34, 579–581. [Google Scholar] [CrossRef] [PubMed]
  154. Satoh, A.; Miura, T.; Satoh, K.; Masamune, A.; Yamagiwa, T.; Sakai, Y.; Shibuya, K.; Takeda, K.; Kaku, M.; Shimosegawa, T. Human leukocyte antigen-DR expression on peripheral monocytes as a predictive marker of sepsis during acute pancreatitis. Pancreas 2002, 25, 245–250. [Google Scholar] [CrossRef]
  155. Kylanpaa, M.L.; Mentula, P.; Kemppainen, E.; Puolakkainen, P.; Aittomaki, S.; Silvennoinen, O.; Haapiainen, R.; Repo, H. Monocyte anergy is present in patients with severe acute pancreatitis and is significantly alleviated by granulocyte-macrophage colony-stimulating factor and interferon-gamma in vitro. Pancreas 2005, 31, 23–27. [Google Scholar] [CrossRef] [PubMed]
  156. Ho, Y.P.; Chiu, C.T.; Sheen, I.S.; Tseng, S.C.; Lai, P.C.; Ho, S.Y.; Chen, W.T.; Lin, T.N.; Lin, C.Y. Tumor necrosis factor-alpha and interleukin-10 contribute to immunoparalysis in patients with acute pancreatitis. Hum. Immunol. 2011, 72, 18–23. [Google Scholar] [CrossRef] [PubMed]
  157. Lin, Z.Q.; Guo, J.; Xia, Q.; Yang, X.N.; Huang, W.; Huang, Z.W.; Xue, P. Human leukocyte antigen-DR expression on peripheral monocytes may be an early marker for secondary infection in severe acute pancreatitis. Hepatogastroenterology 2013, 60, 1896–1902. [Google Scholar] [PubMed]
  158. Gotzinger, P.; Sautner, T.; Spittler, A.; Barlan, M.; Wamser, P.; Roth, E.; Jakesz, R.; Fugger, R. Severe acute pancreatitis causes alterations in HLA-DR and CD14 expression on peripheral blood monocytes independently of surgical treatment. Eur. J. Surg. 2000, 166, 628–632. [Google Scholar] [PubMed]
  159. Wolk, K.; Hoflich, C.; Zuckermann-Becker, H.; Docke, W.D.; Volk, H.D.; Sabat, R. Reduced monocyte CD86 expression in postinflammatory immunodeficiency. Crit. Care Med. 2007, 35, 458–467. [Google Scholar] [CrossRef] [PubMed]
  160. Zhang, Y.; Wang, C.; Yu, M.; Zhao, X.; Du, J.; Li, Y.; Jing, H.; Dong, Z.; Kou, J.; Bi, Y.; et al. Neutrophil extracellular traps induced by activated platelets contribute to procoagulant activity in patients with colorectal cancer. Thromb. Res. 2019, 180, 87–97. [Google Scholar] [CrossRef] [PubMed]
  161. Ferat-Osorio, E.; Wong-Baeza, I.; Esquivel-Callejas, N.; Figueroa-Figueroa, S.; Duarte-Rojo, A.; Guzman-Valdivia-Gomez, G.; Rodea-Rosas, H.; Torres-Gonzalez, R.; Sanchez-Fernandez, P.; Arriaga-Pizano, L.; et al. Triggering receptor expressed on myeloid cells-1 expression on monocytes is associated with inflammation but not with infection in acute pancreatitis. Crit. Care 2009, 13, R69. [Google Scholar] [CrossRef] [PubMed]
  162. Hue, S.; Beldi-Ferchiou, A.; Bendib, I.; Surenaud, M.; Fourati, S.; Frapard, T.; Rivoal, S.; Razazi, K.; Carteaux, G.; Delfau-Larue, M.H.; et al. Uncontrolled innate and impaired adaptive immune responses in patients with COVID-19 acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2020, 202, 1509–1519. [Google Scholar] [CrossRef] [PubMed]
  163. Peruzzi, B.; Bencini, S.; Capone, M.; Mazzoni, A.; Maggi, L.; Salvati, L.; Vanni, A.; Orazzini, C.; Nozzoli, C.; Morettini, A.; et al. Quantitative and qualitative alterations of circulating myeloid cells and plasmacytoid DC in SARS-CoV-2 infection. Immunology 2020, 161, 345–353. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, F.; Hou, H.; Yao, Y.; Wu, S.; Huang, M.; Ran, X.; Zhou, H.; Liu, Z.; Sun, Z. Systemically comparing host immunity between survived and deceased COVID-19 patients. Cell Mol. Immunol. 2020, 17, 875–877. [Google Scholar] [CrossRef] [PubMed]
  165. Pfortmueller, C.A.; Meisel, C.; Fux, M.; Schefold, J.C. Assessment of immune organ dysfunction in critical illness: Utility of innate immune response markers. Intensive Care Med. Exp. 2017, 5, 49. [Google Scholar] [CrossRef] [PubMed]
  166. Boomer, J.S.; To, K.; Chang, K.C.; Takasu, O.; Osborne, D.F.; Walton, A.H.; Bricker, T.L.; Jarman, S.D., 2nd; Kreisel, D.; Krupnick, A.S.; et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 2011, 306, 2594–2605. [Google Scholar] [CrossRef] [PubMed]
  167. Monneret, G.; Lepape, A.; Venet, F. A dynamic view of mHLA-DR expression in management of severe septic patients. Crit. Care 2011, 15, 198. [Google Scholar] [CrossRef] [Green Version]
  168. Wu, L.M.; Sankaran, S.J.; Plank, L.D.; Windsor, J.A.; Petrov, M.S. Meta-analysis of gut barrier dysfunction in patients with acute pancreatitis. Br. J. Surg. 2014, 101, 1644–1656. [Google Scholar] [CrossRef]
  169. Qin, Y.; Pinhu, L.; You, Y.; Sooranna, S.; Huang, Z.; Zhou, X.; Yin, Y.; Song, S. The role of Fas expression on the occurrence of immunosuppression in severe acute pancreatitis. Dig. Dis. Sci. 2013, 58, 3300–3307. [Google Scholar] [CrossRef] [PubMed]
  170. Sharma, D.; Jakkampudi, A.; Reddy, R.; Reddy, P.B.; Patil, A.; Murthy, H.V.V.; Rao, G.V.; Reddy, D.N.; Talukdar, R. Association of systemic inflammatory and anti-inflammatory responses with adverse outcomes in acute pancreatitis: Preliminary results of an ongoing study. Dig. Dis. Sci. 2017, 62, 3468–3478. [Google Scholar] [CrossRef]
  171. Yu, Z.X.; Chen, X.C.; Zhang, B.Y.; Liu, N.; Gu, Q. Association between HLA-DR expression and multidrug-resistant infection in patients with severe acute pancreatitis. Curr. Med. Sci. 2018, 38, 449–454. [Google Scholar] [CrossRef] [PubMed]
  172. Giamarellos-Bourboulis, E.J. Complex immune deregulation in severe COVID-19: More than a mechanism of pathogenesis. EBioMedicine 2021, 73, 103673. [Google Scholar] [CrossRef]
  173. Loftus, T.J.; Ungaro, R.; Dirain, M.; Efron, P.A.; Mazer, M.B.; Remy, K.E.; Hotchkiss, R.S.; Zhong, L.; Bacher, R.; Starostik, P.; et al. Overlapping but disparate inflammatory and immunosuppressive responses to SARS-CoV-2 and bacterial sepsis: An immunological time course analysis. Front. Immunol. 2021, 12, 792448. [Google Scholar] [CrossRef]
  174. Ding, L.; Wan, M.; Wang, D.; Cao, H.; Wang, H.; Gao, P. Myeloid-derived suppressor cells in patients with acute pancreatitis with increased inhibitory function. Front. Immunol. 2022, 13, 840620. [Google Scholar] [CrossRef] [PubMed]
  175. Grassi, G.; Notari, S.; Gili, S.; Bordoni, V.; Casetti, R.; Cimini, E.; Tartaglia, E.; Mariotti, D.; Agrati, C.; Sacchi, A. Myeloid-derived suppressor cells in COVID-19: The paradox of good. Front. Immunol. 2022, 13, 842949. [Google Scholar] [CrossRef]
  176. Falck-Jones, S.; Österberg, B.; Smed-Sörensen, A. Respiratory and systemic monocytes, dendritic cells and myeloid-derived suppressor cells in COVID-19: Implications for disease severity. J. Intern. Med. 2022, 293, 130–143. [Google Scholar] [CrossRef]
  177. Kylänpää, M.L.; Repo, H.; Puolakkainen, P.A. Inflammation and immunosuppression in severe acute pancreatitis. World J. Gastroenterol. 2010, 16, 2867–2872. [Google Scholar] [CrossRef] [PubMed]
  178. Monneret, G.; Venet, F.; Meisel, C.; Schefold, J.C. Assessment of monocytic HLA-DR expression in ICU patients: Analytical issues for multicentric flow cytometry studies. Crit. Care 2010, 14, 432. [Google Scholar] [CrossRef] [PubMed]
  179. De Waele, J.; Vogelaers, D.; Decruyenaere, J.; De Vos, M.; Colardyn, F. Infectious complications of acute pancreatitis. Acta Clin. Belg. 2004, 59, 90–96. [Google Scholar] [CrossRef] [PubMed]
  180. Leppäniemi, A.; Tolonen, M.; Tarasconi, A.; Segovia-Lohse, H.; Gamberini, E.; Kirkpatrick, A.W.; Ball, C.G.; Parry, N.; Sartelli, M.; Wolbrink, D.; et al. 2019 WSES guidelines for the management of severe acute pancreatitis. World J. Emerg. Surg. 2019, 14, 27. [Google Scholar] [CrossRef]
  181. Estella, Á.; Vidal-Cortés, P.; Rodríguez, A.; Andaluz Ojeda, D.; Martín-Loeches, I.; Díaz, E.; Suberviola, B.; Gracia Arnillas, M.P.; Catalán González, M.; Álvarez-Lerma, F.; et al. Management of infectious complications associated with coronavirus infection in severe patients admitted to ICU. Med. Intensiv. 2021, 45, 485–500. [Google Scholar] [CrossRef] [PubMed]
  182. Dickel, S.; Grimm, C.; Amschler, K.; Schnitzler, S.U.; Schanz, J.; Moerer, O.; Payen, D.; Tampe, B.; Winkler, M.S. Case report: Interferon-γ restores monocytic human leukocyte antigen receptor (mHLA-DR) in severe COVID-19 with acquired immunosuppression syndrome. Front. Immunol. 2021, 12, 645124. [Google Scholar] [CrossRef]
  183. van Laarhoven, A.; Kurver, L.; Overheul, G.J.; Kooistra, E.J.; Abdo, W.F.; van Crevel, R.; Duivenvoorden, R.; Kox, M.; Ten Oever, J.; Schouten, J.; et al. Interferon gamma immunotherapy in five critically ill COVID-19 patients with impaired cellular immunity: A case series. Med (N. Y.) 2021, 2, 1163–1170.e2. [Google Scholar] [CrossRef] [PubMed]
  184. Nguyen, L.S.; Ait Hamou, Z.; Gastli, N.; Chapuis, N.; Pène, F. Potential role for interferon gamma in the treatment of recurrent ventilator-acquired pneumonia in patients with COVID-19: A hypothesis. Intensive Care Med. 2021, 47, 619–621. [Google Scholar] [CrossRef] [PubMed]
  185. Laterre, P.F.; François, B.; Collienne, C.; Hantson, P.; Jeannet, R.; Remy, K.E.; Hotchkiss, R.S. Association of interleukin 7 immunotherapy with lymphocyte counts among patients with severe coronavirus disease 2019 (COVID-19). JAMA Netw. Open 2020, 3, e2016485. [Google Scholar] [CrossRef] [PubMed]
  186. Ke, L.; Zhou, J.; Mao, W.; Chen, T.; Zhu, Y.; Pan, X.; Mei, H.; Singh, V.; Buxbaum, J.; Doig, G.; et al. Immune enhancement in patients with predicted severe acute necrotising pancreatitis: A multicentre double-blind randomised controlled trial. Intensive Care Med. 2022, 48, 899–909. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathogenesis of inflammation in AP and COVID-19. Acute pathological insults of SARS-CoV-2 infection and pancreatic acinar cell injury elicit local inflammation mediated by cytokines, unsaturated fatty acids (UFAs), damage-associated molecular patterns (DAMPs), and/or pathogen-associated molecular patterns (PAMPs). The pro-inflammatory reaction induces an anti-inflammatory response to restrict inflammation. When the pro-/anti-inflammation is unbalanced and dysregulated, systemic inflammatory response syndrome (SIRS) or compensatory anti-inflammatory response syndrome (CARS) occurs. During SIRS, monocytes are hyperactivated in response to high levels of pro-inflammatory cytokines and chemokines. In contrast, during CARS, monocytes are deactivated, exhibit reduced mHLA-DR expression, and are incapable of presenting antigens to activate CD4+ T lymphocytes.
Figure 1. Pathogenesis of inflammation in AP and COVID-19. Acute pathological insults of SARS-CoV-2 infection and pancreatic acinar cell injury elicit local inflammation mediated by cytokines, unsaturated fatty acids (UFAs), damage-associated molecular patterns (DAMPs), and/or pathogen-associated molecular patterns (PAMPs). The pro-inflammatory reaction induces an anti-inflammatory response to restrict inflammation. When the pro-/anti-inflammation is unbalanced and dysregulated, systemic inflammatory response syndrome (SIRS) or compensatory anti-inflammatory response syndrome (CARS) occurs. During SIRS, monocytes are hyperactivated in response to high levels of pro-inflammatory cytokines and chemokines. In contrast, during CARS, monocytes are deactivated, exhibit reduced mHLA-DR expression, and are incapable of presenting antigens to activate CD4+ T lymphocytes.
Ijms 24 03246 g001
Figure 2. Measurement of mHLA-DR expression. (A) Measurement of mHLA-DR expression and requirements for sample handling procedures [97,98,99,100,101]. (B) Relationship of units of mHLA-DR expression to different measurement methods.
Figure 2. Measurement of mHLA-DR expression. (A) Measurement of mHLA-DR expression and requirements for sample handling procedures [97,98,99,100,101]. (B) Relationship of units of mHLA-DR expression to different measurement methods.
Ijms 24 03246 g002
Figure 3. Regulation of mHLA-DR expression. Figure referenced from [105,111]. The transcription of HLA-DR is tightly regulated by a set of cis-acting regulatory promoter elements and transcription factors. Class II transactivator (CIITA) is the master transcriptional regulator. The α- and β-chains of HLA-DR assemble in the endoplasmic reticulum (ER) and then bind with the invariant chain (Ii). The Ii–HLA-DR complexes transport through Golgi complex to the MHC class II compartment (MIIC), directly or via the internalization of the plasma membrane. Ii is degraded into class II-associated Ii peptide (CLIP) via members of cathepsin family. In the aid of chaperone HLA-DM, CLIP is exchanged for antigen peptide. Peptide-HLA-DR complexes are then transported to the plasma membrane for further T cell activation. Interfering with the expression and activity of CIITA, Ii, cathepsins, HLA-DM, as well as the associated vesicle traffic, all result in alteration of the mHLA-DR expression.
Figure 3. Regulation of mHLA-DR expression. Figure referenced from [105,111]. The transcription of HLA-DR is tightly regulated by a set of cis-acting regulatory promoter elements and transcription factors. Class II transactivator (CIITA) is the master transcriptional regulator. The α- and β-chains of HLA-DR assemble in the endoplasmic reticulum (ER) and then bind with the invariant chain (Ii). The Ii–HLA-DR complexes transport through Golgi complex to the MHC class II compartment (MIIC), directly or via the internalization of the plasma membrane. Ii is degraded into class II-associated Ii peptide (CLIP) via members of cathepsin family. In the aid of chaperone HLA-DM, CLIP is exchanged for antigen peptide. Peptide-HLA-DR complexes are then transported to the plasma membrane for further T cell activation. Interfering with the expression and activity of CIITA, Ii, cathepsins, HLA-DM, as well as the associated vesicle traffic, all result in alteration of the mHLA-DR expression.
Ijms 24 03246 g003
Figure 4. Immune response in AP and COVID-19. Figure referenced from [121,122]. Pro- and anti-inflammatory response are both activated after acute insults of either AP or COVID-19. The initial generalized inflammation is individually heterogeneous. Patients with less intense generalized inflammatory response may survive and restore the immune balance. When inflammation markedly outpaces anti-inflammation, monocytes are hyperactivated, leading to increased systemic release of pro-inflammatory cytokines and resulting in systemic inflammatory response syndrome (SIRS). This cytokine storm and hyperinflammation is associated with multiple organ failure (MOF) and mortality in AP and COVID-19. Conversely, compensatory anti-inflammatory response syndrome (CARS) happens when the anti-inflammatory response is overwhelming. mHLA-DR is an indicator of this, and expression below 15,000 AB/c or 80% characterizes immunosuppression and below 5000 AB/c or 30% characterizes immunoparalysis. In addition to pronouncedly reduced mHLA-DR expression, monocytes are deactivated with TNF-α production upon lipopolysaccharide (LPS) stimulation in CARS. Lymphocytes are depleted, accompanied by a massive release of anti-inflammatory cytokine. This dysregulated and persistent immunosuppression contributes to MOF, death, and infections.
Figure 4. Immune response in AP and COVID-19. Figure referenced from [121,122]. Pro- and anti-inflammatory response are both activated after acute insults of either AP or COVID-19. The initial generalized inflammation is individually heterogeneous. Patients with less intense generalized inflammatory response may survive and restore the immune balance. When inflammation markedly outpaces anti-inflammation, monocytes are hyperactivated, leading to increased systemic release of pro-inflammatory cytokines and resulting in systemic inflammatory response syndrome (SIRS). This cytokine storm and hyperinflammation is associated with multiple organ failure (MOF) and mortality in AP and COVID-19. Conversely, compensatory anti-inflammatory response syndrome (CARS) happens when the anti-inflammatory response is overwhelming. mHLA-DR is an indicator of this, and expression below 15,000 AB/c or 80% characterizes immunosuppression and below 5000 AB/c or 30% characterizes immunoparalysis. In addition to pronouncedly reduced mHLA-DR expression, monocytes are deactivated with TNF-α production upon lipopolysaccharide (LPS) stimulation in CARS. Lymphocytes are depleted, accompanied by a massive release of anti-inflammatory cytokine. This dysregulated and persistent immunosuppression contributes to MOF, death, and infections.
Ijms 24 03246 g004
Table 1. Cytokine Modulation of HLA-DR Expression.
Table 1. Cytokine Modulation of HLA-DR Expression.
CytokinesHLA-DR ExpressionRegulatory MechanismsReferences
IL-10Downregulation of CIITA; Altering vesicular traffic of HLA-DR in exocytosis and recycling[112,113]
TGF-βInhibition of CIITA and downregulation of HLA-DR transcription[114,115]
IFN-βDownregulation of CIITA[116]
IFN-γPromotion of HLA-DR and CD74 transcription[117,118]
GM-CSFPromoting exocytosis and reducing internalization[119]
TNF-α, IL-1Boosting biosynthesis and stability of HLA-DR increasing half-life from about 10 h to over 100 h[120]
IL-4Upregulation of CIITA[113]
Abbreviations: IL, interleukin; CIITA, class II transactivator; TGF-β, transforming growth factor-beta; IFN, interferon; GM-CSF, granulocyte-macrophage colony-stimulating factor; TNF-α, tumor necrosis factor-alpha.
Table 2. Predictive Values of mHLA-DR for severity and mortality in AP and COVID-19.
Table 2. Predictive Values of mHLA-DR for severity and mortality in AP and COVID-19.
DiseasePredictionSample Size (Incidents/Total)Measuring TimeCut-Off ValueAUCSensitivitySpecificityOthers
AP [149]MAP from MSAP/SAP27/50Admission<2274 MFI0.80570.4%82.6%Combined with classical monocyte proportions (AUC, 0.862)
SAP from MSAP9/23<1094.5 MFI0.69085.7%55.6%-
AP [150]SAP19/58Admission<50.8%0.72872%72%-
Day 2<43.35%0.80084%80%-
Day 5<60.8%0.87782%78%-
AP [151] Organ failure29/310Admission<78% 0.7883%72%-
<38 RFU0.8169%84%-
AP [152] Mortality7/25Day 10<52.3%0.94494.4%85.7%-
COVID-19 [136]Critical COVID-199/32Days 0–3<81.55%0.961100.00%80.00%-
COVID-19 [139]Severe COVID-1948/97Admission<143 MFI0.989.6%81.6%Independent predictor of COVID-19 severity (OR = 0.976, 95% CI: 0.955–0.997)
COVID-19 [134]Mortality35/124Days 0–3<11,312 AB/c0.6474%54%-
Days 7–10<4672 AB/c0.8575%86%-
COVID-19 [153] Mortality1/12Admission<270.56 cells/mL0.875100.0%87.5%-
Abbreviations: AUC, area under the receiver operating characteristic curve; MAP, mild acute pancreatitis; (M)SAP, (moderately) severe acute pancreatitis; MFI, mean fluorescence intensity; RFU, relative fluorescence unit; OR, odds ratio; CI, confidence interval.
Table 3. Predictive values of mHLA-DR for septic complications in AP and COVID-19.
Table 3. Predictive values of mHLA-DR for septic complications in AP and COVID-19.
DiseasePredictionSample Size (Incidents/Total)Measuring TimeCut-Off ValueAUCSensitivitySpecificityOthers
AP [154] Sepsis6/64Admission<60%-100%91.3%Superior to Ranson’s score and APACHE II score
Day 7-100%93.2%-
Day 14-100%98.2%-
AP [128]Septic complications11/74Day 7<40%-73%94%-
Day 10-82%98%-
Day 14-100%100%-
AP [152]Septic complications6/25Day 10<58%0.92676.5%100%Comparable to Ranson and APACHE II scores and better than CRP levels (AUC, 0.841, 0.869, and 0.460, respectively)
AP [157]Secondary infection11/40Admission<35.8%0.83781.8%82.8%-
COVID-19 [134]Secondary infection38/124Days 0–3<10,523 AB/c 0.7076%60%-
Days 7–10<6804 AB/c0.6277%52%-
Abbreviations: AUC, area under the receiver operating characteristic curve; APACHE, Acute Physiology and Chronic Health Evaluation; CRP, C-reactive protein.
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

Liu, S.; Luo, W.; Szatmary, P.; Zhang, X.; Lin, J.-W.; Chen, L.; Liu, D.; Sutton, R.; Xia, Q.; Jin, T.; et al. Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19. Int. J. Mol. Sci. 2023, 24, 3246. https://doi.org/10.3390/ijms24043246

AMA Style

Liu S, Luo W, Szatmary P, Zhang X, Lin J-W, Chen L, Liu D, Sutton R, Xia Q, Jin T, et al. Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19. International Journal of Molecular Sciences. 2023; 24(4):3246. https://doi.org/10.3390/ijms24043246

Chicago/Turabian Style

Liu, Shiyu, Wenjuan Luo, Peter Szatmary, Xiaoying Zhang, Jing-Wen Lin, Lu Chen, Dan Liu, Robert Sutton, Qing Xia, Tao Jin, and et al. 2023. "Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19" International Journal of Molecular Sciences 24, no. 4: 3246. https://doi.org/10.3390/ijms24043246

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