Hostname: page-component-6bf8c574d5-gr6zb Total loading time: 0 Render date: 2025-02-20T07:21:46.718Z Has data issue: false hasContentIssue false

Overview of the cellular and immune mechanisms involved in acute pancreatitis: In search of new prognosis biomarkers

Published online by Cambridge University Press:  06 January 2025

Alexandra Mititelu
Affiliation:
2nd Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
Alina Grama*
Affiliation:
2nd Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
Marius-Cosmin Colceriu
Affiliation:
2nd Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
Tudor L. Pop
Affiliation:
2nd Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
*
Corresponding author: Alina Grama; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Acute pancreatitis (AP) is an acute-onset gastrointestinal disease characterized by a significant inflammation of the pancreas. Most of the time, AP does not leave substantial changes in the pancreas after the resolution of the symptoms but the severe forms are associated with local or systemic complications. The pathogenesis of AP has long been investigated and, lately, the importance of intracellular mechanisms and the immune system has been described. The initial modifications in AP take place in the acinar cell. There are multiple mechanisms by which cellular homeostasis is impaired, one of the most important being calcium overload. Necrotic pancreatic cells initiate the inflammatory response by secreting inflammatory mediators and attracting immune cells. From this point on, the inflammation is sustained by the involvement of innate and adaptive immune systems. Multiple studies have demonstrated the importance of the first 48 h for identifying patients at risk for developing severe forms. For this reason, there is a need to find new, easy-to-use and reliable markers for accurate predictions of these forms. This review provides an overview of the main pathogenetic mechanisms involved in AP development and the most promising biomarkers for severity stratification.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press

Introduction

Acute pancreatitis (AP) is an important cause of hospitalization and a burden for the healthcare system through the resources needed for the management and follow-up of these patients (Refs Reference Toh, Phillips and Johnson1, Reference Singla2). It is characterized by the acute inflammation of the pancreas that can evolve into multiple organ failure and, consequently, have significant morbidity and mortality (Refs Reference Peery3, Reference Sigounas4). In recent years, a progressive increase in the incidence of AP has been observed, both in the adult and paediatric populations, up to 74 cases per 100,000 and 13 cases per 100,000, respectively (Refs Reference Iannuzzi5, Reference Morinville, Barmada and Lowe6). The reason for this increase is not understood but it seems to be determined by increased recognition and environmental and lifestyle changes (Ref. Reference Morinville, Barmada and Lowe6).

The diagnosis of AP is currently established according to the revised Atlanta classification and it requires the fulfilment of two of the following three criteria: abdominal pain characteristic for AP, serum lipase or amylase values over three times the upper normal limit and imaging findings characteristic for AP on abdominal ultrasound, computed tomography or magnetic resonance imaging (Ref. Reference Banks7).

The revised Atlanta classification also establishes the severity of the AP, which recognizes three AP forms: mild AP, in which local or systemic complications are absent; moderate AP, including cases with local complications like pancreatic/peripancreatic collections and/or organ dysfunction that resolves in <48 h; and severe forms including patients with organ dysfunction that persists for more than 48 h, many of them developing multiple organ dysfunction syndrome (MODS) (Ref. Reference Banks7).

AP has different aetiology between adults and children, but regardless of the triggering factor, the pathogenetic mechanisms are the same (Refs Reference Wang8, Reference Suzuki, Sai and Shimizu9). These mechanisms have long been investigated but many aspects remain poorly understood. The initial modification appears in the acinar cell. Several changes impair cellular homeostasis, one of the most studied being premature trypsinogen activation. However, in recent years, other mechanisms like intracellular calcium overload or transcription factor activation have been demonstrated to be very important (Refs Reference Leung and Ip10, Reference Logsdon and Ji11). Destructed acinar cells initiate the inflammatory response by attracting immune cells, such as neutrophils and macrophages. Recently, the involvement of the adaptive immune system has been demonstrated as an essential player in the augmentation and diminution of inflammation (Refs Reference Logsdon and Ji11, Reference Weber and Adler12). Numerous unanswered questions remain regarding the immune dysregulation that differentiates between a local limited form and a severe one characterized by multiorgan involvement.

As stated before, AP is a significant cause of mortality, especially the severe form in which it reaches up to 50% (Ref. Reference Banks7). For this reason, several scoring systems have been studied for the prognosis of severe forms, but none showed reliable predictive values and are difficult to apply (Ref. Reference Sigounas4). The problem is even more significant if we look at paediatric patients, for which these scores cannot be used (Ref. Reference Suzuki, Sai and Shimizu9). These gaps in understanding and managing AP have led to the need to identify new, more reliable makers for the prognosis values and the possible therapeutic purpose.

This review discusses the major mechanisms involved in initiating and progressing AP, intracellular changes and immune system involvement, as well as the most promising markers that could serve to predict the severe forms and even as therapeutic targets.

Pathogenetic mechanisms of acute pancreatitis

Cellular injury

For many years, the central AP mechanism was considered the premature activation of trypsin and autodigestion of the acinar cells. However, it is now clear that several mechanisms produce acinar cell dysfunction and inflammation initiation (Ref. Reference Logsdon and Ji11). The main mechanisms involved are premature activation of trypsin, colocalization of zymogen granules with lysosomes, calcium signalling and activation of the transcription factor nuclear factor-κB (NF-κB) (Ref. Reference Weber and Adler12). Figure 1 show the main cellular mechanisms.

Figure 1. Multiple cellular mechanisms determine apoptosis/necrosis of the acinar cell. Calcium overload is the central mechanism that activates a cascade of events. The intracellular calcium comes from increased ER release and the extracellular space influx. This calcium overload impaired cellular homeostasis by several mechanisms: activation of the nuclear factor κB (NF-κB) pathway and cytokine synthesis; mitochondrial dysfunction and decreased ATP production followed by cellular death; trypsinogen activation and autodigestion followed by cellular destruction; colocalization of zymogens and lysosomes, ductal cell dysfunction. All these processes determine the destruction of acinar cells through necrosis, apoptosis or pyroptosis.

Premature trypsinogen activation

The central role of trypsin activation in the pathogenesis of AP has long been demonstrated, but the mechanisms by which trypsinogen gets activated are not entirely understood. The most substantial evidence supporting the importance of trypsin activation comes from the hereditary forms of pancreatitis. Mutations in the genes encoding cationic trypsinogen (PRSS1) determine an increased susceptibility to acute and chronic pancreatitis by making molecules more susceptible to activation. Similarly, the mutations in the gene encoding the serine protease inhibitor (SPINK1) cause an autosomal recessive susceptibility by secreting an inactive molecule (Refs Reference Dixit13, Reference Watanabe, Kudo and Strober14). Additional support for this hypothesis comes from studies with experimental models of pancreatitis. In a study that used knockout mice lacking trypsinogen isoform-7, there was no intra-acinar trypsinogen activation, but there was evidence of NF-κB activation, and systemic inflammation was not altered (Refs Reference Dixit13, Reference Dawra15). These findings concluded that trypsinogen activation could be an important factor in initiating acinar injury and inflammation cascade from AP but it is not mandatory for the appearance of the disease; other mechanisms determine cellular damage and inflammatory response (Refs Reference Dixit13, Reference Watanabe, Kudo and Strober14).

Colocalization of zymogens with lysosomes

Various pancreatic injuries like trauma, ductal obstruction or alcohol can determine the fusion between the lysosome and zymogen granules inside the acinar cell. This process is called colocalization, and in these vacuoles, cathepsin B, a lysosomal enzyme, determines the activation of trypsinogen to trypsin. After the enzymes are released from the vacuoles, trypsin causes autodigestion, but cathepsin B also plays an important role in cellular destruction. Experimental studies demonstrated that small amounts of cytosol cathepsin B initiate cell death by apoptosis. In contrast, large amounts shift the pathway towards necrosis, thus being an important molecule in the initial phases of AP (Refs Reference Dixit13, Reference Lee and Papachristou16Reference Talukdar18).

Calcium signalling

Usually, intracellular calcium handling is an important mechanism for the function of the acinar cell. Calcium release from endoplasmic reticulum (ER) is associated with zymogen exocytosis in the apical pole and stimulates ATP production in the mitochondria. This increase in cytosolic levels of calcium is transient and, in physiological conditions, the levels rapidly decreased by two ATP-dependent calcium channels: the smooth-ER calcium channel, which moves back the calcium into the ER (SERCA- ssarcoplasmic/endoplasmic reticulum Ca2+ ATPase), and the membrane calcium channel, which removes calcium from the cell (PMCA - plasma membrane Ca2+ ATPase) (Refs Reference Lee and Papachristou16, Reference Wen19). Alcohol and bile acids have been demonstrated to disrupt intracellular calcium homeostasis by increasing calcium levels from the ER store and, afterwards, entering extracellular calcium (Refs Reference Weber and Adler12, Reference Lee and Papachristou16, Reference Voronina20). Furthermore, other aetiologies have been associated with the calcium signalling mechanism, like medication (asparaginase) and post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis, making it one of the most important pathways for cellular injury (Refs Reference Pallagi21, Reference Peng22). The build-up of calcium levels in the acinar cell triggers several important mechanisms that eventually lead to cellular death: trypsinogen auto-activation, vacuole formation, cytoskeletal damage, NF-κB activation and maybe, most importantly, mitochondrial dysfunction. The calcium overload results in a loss of membrane potential to produce ATP, accentuating calcium accumulation by decreasing the function of ATP-dependent channels SERCA and PMCA (Refs Reference Weber and Adler12, Reference Lee and Papachristou16, Reference Raraty23Reference Raraty25). All this evidence suggests that increased intracellular calcium has a central role in the initiating phases of AP. This observation led to the search for potential therapeutic targets that could prevent calcium overload, some of which had encouraged initial results (Ref. Reference Pallagi21).

Autophagy and endoplasmic reticulum stress

Autophagy is an essential cytoprotective process through which cytoplasmatic proteins and organelles are degraded and recycled. Targeted molecules are entrapped into autophagic vacuoles, which merge with lysosomes and are digested by proteolytic enzymes. In AP, autophagy is impaired, resulting in ER stress, premature trypsinogen activation and mitochondrial dysfunction (Refs Reference Watanabe, Kudo and Strober14, Reference Lee and Papachristou16).

ER stress means the accumulation of misfolded proteins inside the ER that happens when the normal function of the ER is overwhelmed and protein elimination is impaired. In AP, ER stress is determined by an increased demand for protein production and defects in recycling proteins (Refs Reference Lee and Papachristou16, Reference Biczo24, Reference Wu26).

Ductal cell dysfunction

The most investigated association between ductal cell involvement in AP is with the cystic fibrosis transmembrane regulator mutations that cause increased susceptibility to developing the disease by reduced secretion (Ref. Reference Lee and Papachristou16). Secretion of bicarbonate from the ductal cells is an essential mechanism for the neutralization of acidic content produced by acinar cells. It is regulated by pH-sensing mechanisms and membrane transporters (Refs Reference Grapin-Botton27, Reference Hegyi28). Recent studies regarding ductal cell function in AP revealed an increased secretion in the initial phase, followed by a reduction afterwards, which seems to be a protective mechanism intended to wash out the toxins (Ref. Reference Venglovecz, Rakonczay and Hegyi29). Another mechanism of ductal damage is increased pressure caused by gallstone obstruction or contrast injection during ERCP, which can activate the pathological calcium signalling pathway in the acinar cells. Other modifications are fluid stasis, which promotes premature enzyme activation, intraductal acidification and ductal cell exposure to bile acids (Ref. Reference Lee and Papachristou16).

NF-κB activation

NF-κB is a transcription factor that regulates several genes involved in immune and inflammatory response by increasing the production of cytokines and chemokines and the activation of innate immune cells and inflammatory lymphocyte T cells (Refs Reference Liu30, Reference Pai and Thomas31). The involvement of NF-κB in acute pancreatitis comes from experimental studies that demonstrated reduced severity of pancreatitis in a model with NF-κB deletion. The exact activation mechanisms are not fully understood but evidence suggests calcium signalling and the production of reactive oxygen species are the most important ones. NF-κB activation in AP determines cytokine synthesis and initiation of local inflammatory response through transcriptional induction of proinflammatory cytokines, chemokines and other inflammatory mediators like inflammasomes (Refs Reference Liu30, Reference Jakkampudi32).

MicroRNAs

MicroRNAs (miRNAs) are single-stranded nucleotides that belong to the non-coding RNA class. These nucleotides play an important role in regulating gene expression and, thus, are involved in cell growth, development and death (Refs Reference O’Brien33, Reference Yang34). The biogenesis of miRNAs is a complex process. Typically, they bind to a complementary sequence in the 3′-untranslated region of messenger RNA (mRNA), causing mRNA silencing or degradation. A single miRNA can regulate the expression of hundreds of genes (Ref. Reference Bartel35). Recently, there has been increasing evidence of the functional role of miRNAs in acinar cell injury, inflammatory response and distal organ involvement (Ref. Reference Yang34). The first evidence to support miRNAs in AP came from studies on animal models that revealed significant upregulation of miR-135a and miR-22 in the pancreatic tissue (Ref. Reference Qin36). Afterwards, many miRNAs, like miR-29a and miR-21-5p, seem to be upregulated in pancreatic acinar cells during AP and induce cellular death through apoptosis, necrosis or pyroptosis (Refs Reference Ma37, Reference Gao38). Furthermore, miRNAs participate in the immune cell regulation in the progression of AP by interacting and upregulating macrophages, mononuclear cells and even T lymphocytes (Refs Reference Yu, Odenthal and Fries39Reference Wang41). miRNAs seem to be involved in AP-associated lung injury, miR-21 being upregulated in lung tissue during severe AP (Ref. Reference Li42). Due to this evidence and the good stability of these markers, miRNAs could be considered as possible predicting markers for severity stratification (Refs Reference Gao38, Reference Yang43).

Apoptosis and necrosis

The final result of all the above-described changes in the acinar cell is cellular death, which is produced either by apoptosis, necrosis or pyroptosis. While apoptosis is a form of programmed cellular death mediated by caspases, necrosis is a process of self-destruction caused by internal or external factors (Ref. Reference Kang44). In AP, necrosis seems to be the main type of pancreatic acinar cell death and the release of damage-associated molecular pattern molecules (DAMPs) from necrosis is implicated in the inflammatory response. The mechanism leading to one or another form of cellular death is still unknown but may be a crucial factor for disease progression (Refs Reference Raraty23, Reference Kang44).

Pyroptosis

Pyroptosis is a form of cellular death that involves the death of immune cells mediated by inflammasomes secondary to an immune stimulus (Ref. Reference Kang44). This type of cellular death has some features similar to apoptosis and necrosis, like DNA fragmentation and cytoplasmic swelling. Unlike apoptosis, pyroptosis has a proinflammatory effect due to the rapid plasma membrane permeabilization and early membrane rupture, as well as the secretion of proinflammatory cytokines (Refs Reference Kang44, Reference Lamkanfi and Dixit45). Pyroptosis progression is a caspase-dependent process. Both the canonical pathway, which involves caspase-1 activation, as well as the noncanonical pathway, involving caspase-4, -5 and -11, can initiate pyroptosis (Ref. Reference Li46). There is increasing evidence that shows the importance of the inflammasomes and, subsequently, pyroptosis in the progression of inflammation in AP. Therefore, pyroptosis-related molecules could be an independent prognostic marker for AP severity but pyroptosis could also provide a therapeutic target for severe forms in the future (Ref. Reference Al Mamun47).

The pathogenetic modifications are the same after initiating the inflammatory process, regardless of the aetiology. Nevertheless, the trigger may differ between the different aetiologies. The main aetiologies and the triggering mechanisms are summarized in Table 1.

Table 1. The main mechanisms for the initiation of AP associated with the most important causes

Note: AP, acute pancreatitis; CFTR, cystic fibrosis transmembrane regulator; ERCP, endoscopic retrograde cholangiopancreatography; NF-κB, nuclear factor-κB.

Innate immune response

The innate immune system is the first line of defence against pathogens. It is a nonspecific but very rapid protection mechanism without immunological memory. There are some molecules released by the pathogens that are critical in the initialization of the innate immune response: pathogen-associated molecular patterns (PAMPs), lipopolysaccharides or double-stranded ribonucleic acid (RNA). In the innate immune response, many types of cells are implicated: phagocytes (neutrophils and macrophages), dendritic cells (DCs), mast cells and natural killer (NK) cells (Refs Reference Murphy, Travers and Walport53, Reference Marshall54).

In AP, damaged acinar cells release cytokines, chemokines and DAMPs, which determine the rapid recruitment of immune cells in the pancreas. The main cytokines released in the first stages of inflammation or infection are tumour necrosis factor (TNF), interleukin 1 (IL-1) and IL-6 (Refs Reference Marshall54, Reference Hoque55). Figure 2 show the main innate cells involved in AP.

Figure 2. Necrotic acinar cells release proinflammatory cytokines and chemokines. They are crucial in attracting immune cells to the pancreas: neutrophils, macrophages, dendritic cells and natural killer (NK) cells. The neutrophils are the first ones that migrate to the pancreas and determine augmentation of inflammation augmentation through the release of elastase, myeloperoxidase (MPO) and neutrophil extracellular traps (NETs). Proinflammatory cytokines cause activation of resident macrophage as well as migration of blood monocytes. In the pancreas, monocytes differentiate into M1 (proinflammatory secreting macrophages) and M2 (anti-inflammatory secreting macrophages). The M1/M2 ratio imbalance is an important step in the augmentation of inflammation in acute pancreatitis (AP). Nevertheless, the circulating proinflammatory cells activate the resident macrophages from the liver and lung and determine the magnitude of systemic complications. Dendritic cells also migrate in the pancreas tissue and are involved in the secretion of proinflammatory molecules. The NK cells have a bivalent role, either stimulating macrophages and dendritic cell activation or destroying them by cytotoxic effect.

Neutrophils

Neutrophils are the predominant leukocyte subpopulation in the blood, derived from bone marrow haematopoietic stem cells (Ref. Reference Castanheira and Kubes56). It has been demonstrated that neutrophils are an important factor involved not only in infections but also in sterile inflammation (Ref. Reference Peng, Li and Yu57). Thus, neutrophils are one of the first cells recruited in the pancreas by DAMPs or by the chemokines and leukotriene released by the surrounding tissue to respond to DAMPs production. DAMPs include histone, DNA, IL-1α and many others. From the chemokine family, IL-8 (CXCL8) has the most important role in neutrophil chemotaxis and has been demonstrated to be an essential predictive marker for severity (Refs Reference Peng, Li and Yu57Reference Wang59).

The first step of the recruitment is the interaction with the endothelium, which is enhanced by adhesion molecules from the selectin and integrin family, like intercellular adhesion molecule 1 (ICAM1). Another essential molecule for neutrophil migration is metalloproteinase, especially matrix metalloproteinase-9 (MMP-9), which helps by degrading some components of the basement membrane so the neutrophils can reach the pancreatic tissue (Refs. Reference Wang59Reference Wan61). Once infiltrated in the pancreas, neutrophils perform their functions by phagocytosis, degranulation and neutrophil extracellular trap (NET) production. The most important molecules released by neutrophils are elastase, reactive oxygen species (ROS) and myeloperoxidase, which are involved in the activation of trypsinogen, cytokine release and inflammation enhancement (Refs Reference Peng, Li and Yu57, Reference Wang59).

NETs are another way for neutrophils to fight bacterial infections, but they have also been involved in sterile inflammation. NETs contain DNA, histones and cytosolic proteins and they are usually formed once neutrophils are dead (Refs Reference Castanheira and Kubes56, Reference Wang59).

Neutrophils are also involved in the systemic complications that are associated with severe AP, especially with acute lung injury (ALI). The first phase of ALI is the exudative phase, which involves microvascular damage and endothelial barrier dysfunction induced by inflammatory mediators. These changes determine neutrophil recruitment in the pulmonary interstitium and alveolar space that become activated and produce an increased inflammatory response, similar to the effect on the pancreas (Ref. Reference Zhou62).

Although neutrophils play a significant role in exacerbating inflammation, they also contribute to tissue repair. This is achieved by the phagocytosis of cellular debris, but most importantly, by the apoptosis of neutrophils cleared by macrophages. The clearance process activates the secretion of anti-inflammatory cytokines like tissue-repairing cytokines transforming growth factor-β (TGF-β) and IL-10 (Refs Reference Wang59, Reference Soehnlein and Lindbom63).

Macrophages

Macrophages are one of the most important cells involved in the evolution of AP. Pancreatic macrophages include tissue-resident macrophages and migrating macrophages (circulating monocytes) (Ref. Reference Hu64). DAMPs, proinflammatory mediators released by pancreatic injured tissue, necrotic cells like monocyte chemoattractant protein-1 (MCP-1) and TNF-α induce resident macrophage activation and monocyte migration (Ref. Reference Castanheira and Kubes56). After macrophage recruitment, the predominant cytokine environment determines the orientation of macrophage polarization, meaning that there are two distinct subsets: M1, classically activated, and M2, alternatively activated. M1 macrophages induce a Th1-type response by releasing cytokines and chemokines, accentuating the proinflammatory response (Refs Reference Hu64, Reference Mantovani65). Mosser proposed an alternative classification of macrophages based on three homeostatic activities, which are host defence, wound healing and immune regulation. Furthermore, tumour-associated macrophages have also been identified as a separate group extensively studied in the last few years (Refs Reference Mosser and Edwards66, Reference Mamilos67). The process of macrophage differentiation is called plasticity and describes the ability of cells to respond to different microenvironmental influences by displaying diverse functional phenotypes. Unlike other cells, which lose their heterogeneity during maturation, macrophages retain their plasticity and transform according to environmental signals (Refs Reference Mamilos67, Reference Katkar and Ghosh68).

The severity of the pancreatitis episode is correlated with the degree of macrophage activation and the M1 to M2 ratio imbalance; M1 predominance is associated with more severe forms (Refs Reference Peng, Li and Yu57, Reference Hu64, Reference Perides69). The central role of pancreatic macrophages in the evolution of AP is due to the involvement, besides the cytokine secretion, in trypsinogen activation, NF-κB activation and adaptive immune response (Ref. Reference Hu64).

In addition to pancreatic macrophages, peritoneal, liver and alveolar macrophages play an important role in the evolution of AP. The proinflammatory cytokines and enzymes are released in the bloodstream and the peripancreatic area, inducing the activation of peritoneal macrophages to the M1 subtype. The activated peritoneal macrophage can synthesize chemokines and cytokines like IL-1β, TNF-α and IL-8, aggravating the inflammatory response (Refs Reference Hu64, Reference Samanta70).

Kupffer cells are the resident macrophages of the liver and become activated in the evolution of pancreatitis by the cytokines and enzymes that reach the liver through the portal vein. They influence the magnitude of inflammatory systemic response and pulmonary involvement (Refs Reference Hu64, Reference Liu71).

The alveolar macrophages are activated by the proinflammatory cytokines released by the pancreas and the Kupffer cells, which reach the lung through the blood. The resident pulmonary macrophages determine the severity of pulmonary involvement by cytokine and ROS release (Refs Reference Hu64, Reference Gea-Sorlí72, Reference Gea-Sorlí and Closa73).

DCs

DCs link innate and adaptative immune systems by functioning as phagocytes and antigen-presenting cells. In AP, DCs infiltrate the pancreas and increase inflammation by secreting IL-6, TNF-α and MCP-1 (Ref. Reference Wan61). Also, it has been described that acinar cells can transition to DCs and activate CD4+ T cells, thereby accentuating the local inflammation (Refs Reference Peng, Li and Yu57, Reference Xu74). On the other hand, some studies have demonstrated the protective effect of DCs on the pancreas by inducing severe forms in mice depleted of DCs (Refs Reference Peng, Li and Yu57, Reference Bedrosian75).

NK cells

NK cells are a subtype of circulating lymphocyte that has a cytotoxic effect on tumoural and virus-infected cells. The cytotoxic effect is accomplished by inducing cell apoptosis by releasing perforins and lytic proteins from cytoplasmatic granules (Refs Reference Marshall54, Reference Vivier76). Besides this function, NKs are involved in inflammation through cytokine secretion (interferon-γ (IFN-γ), TNF, IL-5, IL-10 and IL-13) and chemokines (CCL3, CCL4 and IL-8) (Refs Reference Peng, Li and Yu57, Reference Zitti and Bryceson77).

NK cells regulate immune response by activating macrophages, DCs and T cells through the cytokine secreted and surface receptors. On the other hand, NK can kill these cells (DCs and macrophages) by cytotoxic effect (Ref. Reference Vivier76).

Regarding the involvement of NK in AP, evidence suggests that the number of NKs in the peripheral blood is decreased compared to the healthy individual. Moreover, in severe forms, there is an even more considerable decrease and this depletion is correlated to the infectious complications in AP (Ref. Reference Peng, Li and Yu57).

Inflammasomes

Inflammasomes are a cluster of multiprotein complexes found in the cytosol involved in the inflammatory response as part of innate immunity (Refs Reference Ferrero-Andrés78, Reference Martinon, Burns and Tschopp79). These complexes have two important components: one is the upstream sensor protein that belongs to the NLR (NOD like receptor) or ALR (AIM2 like receptor) family, and the other one is the adaptor protein, containing a caspase recruitment domain (ASC - apoptosis-associated speck-like protein containing a caspase activating and recruitment domain) (Ref. Reference Ferrero-Andrés78). The most investigated inflammasome from the NLR family is the NLR pyrin domain-containing protein 3 (NLRP3), which is expressed in monocytes, neutrophils, lymphocytes, DCs and epithelial cells (Ref. Reference Ferrero-Andrés78). NLRP3 is activated by both PAMPS and DAMPs. However, in AP, the recognition of DAMPs by the NLRP3 is the first step in its activation and afterwards in the induction of sterile inflammatory response (Ref. Reference Ferrero-Andrés78). Inflammasomes need two different signals for activation. The first signal requires the upregulation of inflammasome mRNA by NF-κB and the second signal begins the activation of pro-caspase-1 to its active form, caspase-1 (Refs Reference Tsomidis, Voumvouraki and Kouroumalis80, Reference Guo, Callaway and Ting81). There are also two pathways for NLRP3 inflammasome action. The canonical pathway activates caspase-1 and subsequently induces the maturation of IL-1β and IL-18, as well as pyroptosis, a form of programmed cell death discussed earlier. In contrast, noncanonical activation of caspase-1 requires the presence of caspase-11 in addition to the NLRP3 inflammasome (Ref. Reference Lamkanfi and Dixit45). NLRP3 inflammasome is involved in AP inflammation by activating IL-1β and IL-18. These two cytokines are secreted by activated macrophages but require activation from the precursor form, pro-IL-1β and pro-IL-18, to the mature activated form (IL-1β and IL-18). There are two steps in the activation process: the first one is the increased expression of inflammasome components and pro-IL-1β by the NF-κB transcription factor and the second one is the proteolytic maturation of the pro-molecules in the presence of NLRP3 inflammasome and caspase-1 (Ref. Reference Glaubitz82). The involvement of NLRP3 in AP pathogenesis is sustained by experimental studies, which showed that the absence of caspase-1, ASC or NLRP3 substantially reduced oedema and inflammation in a pancreatitis mice model (Refs Reference Ferrero-Andrés78, Reference Hoque83). Furthermore, there is also clinical evidence of the involvement of inflammasomes in AP. In a study conducted by Algaba-Chueca et al. (Ref. Reference Algaba-Chueca84), the levels of NLRP3 and derived IL-1β and IL-18 were elevated in the first stages of AP. Another evidence of the importance of these molecules in AP comes from experimental studies that showed decreased severity of AP when NLRP3 inhibitor was used (Ref. Reference Sendler85).

Adaptive immune response

An important role in triggering and shaping the immune response in AP belongs to adaptive immunity (acquired immunity). Adaptive immunity consists of the body’s response against self-antigens and non-self-antigens after presenting the antigenic particles via major histocompatibility complex (MHC) class I and II molecules to T cells. Repeated exposure to an antigen will activate both humoral immunity (antibody-mediated immunity mediated by B lymphocytes) and cellular immunity (mediated by T lymphocytes), and will trigger a hyperinflammatory process (Ref. Reference Stojanovic86). Damage of the acinar cells will determine pancreatic infiltration with innate immune cells (neutrophils and macrophages) and the abnormal activation of adaptive immune cells. In the more severe forms, they will trigger a systemic inflammatory response (SIRS) or MODS. Subsequently, T cells determine a compensatory anti-inflammatory response (CARS) (Ref. Reference Kang44). Figure 3 show the main adaptive immune cell and mechanisms involved.

Figure 3. In acute pancreatitis (AP), blood T cells are reduced due to two mechanisms: the overexpression of Fas and FasL ligands on the surface, leading to premature apoptosis and migration to the inflamed pancreas. The T cells differentiate in the Th1, which augments the inflammatory response through proinflammatory cytokine production, and Th2, which secretes anti-inflammatory cytokines. The imbalance between these two types, with the predominance of Th1 response, is one of the most important factors for AP progression. Another important subtype of T cells is Th22, which reduces the inflammation in the pancreas by secreting interleukin 22 (IL-22). The role of B cells is not very well understood, but it seems they can play a bivalent role, pro- and anti-inflammatory.

T cells

In the initial phases of pancreas injury, there is a reduction in the number of T cells (CD4 and CD8) in the circulation due to an excessive expression of Fas and FasL ligands on the cell surface. Fas (also known as CD25 or Apo-1) is a member of the TNF receptor family, which promotes lymphocyte apoptosis (Refs Reference Sun87, Reference Zhang88). FasL (named CD95L or CD178) is a type-II transmembrane protein expressed on cytotoxic T lymphocytes and NK cells, which also induces programmed cell death (Ref. Reference Pardalis89). This overexpression of Fas ligands results in a significant loss of peripheral T lymphocytes, immunosuppression and bacterial superinfection. The number of peripheral T cells is also reduced due to their migration to the inflamed pancreas (especially CD4 cells infiltrating the inflamed acini). Usually, the reduction is transient in moderate AP (MAP), with the number of cells normalizing after the fifth day of evolution. In the severe forms of AP (SAP), the severity of cellular destruction is correlated with persistent lymphopenia, especially with the reduction of Th cells, which amplifies the complication risk (Ref. Reference Stojanovic86).

Cytokines released secondary to the pancreas injury (IL-2, IL-4 and IL-12) will determine the differentiation of T cells in T helper (Th1, Th2, Th9, Th17, Th22 and follicular T helper) and regulatory T cells (Treg cells).

T helper cells

The Th1 and Th2 subtypes probably play the most important role in adaptive immunity. Th1 cells mainly produce proinflammatory cytokines to mediate cellular immunity, while Th2 cells produce anti-inflammatory cytokines to regulate humoral immunity (Ref. Reference Xiong90). In AP, Th1 cells participated in the early inflammatory response by releasing proinflammatory cytokines, while the Th2 subset secretes anti-inflammatory cytokines to counterbalance excessive inflammatory response (Ref. Reference Rangel-Frausto91).

Th1 (CD4 cells) is essential in the adaptive immune system. Th1 increases cell-mediated response against intracellular bacteria by releasing different mediators (IL-12, IFN-γ and IL-2), activating macrophages, cytotoxic T cells and B lymphocytes. In addition to stimulating the cellular immune system, Th1 promotes the production of IgG, an opsonizing antibody (Ref. Reference Belizário92). IFN-γ stimulates macrophages to produce IL-12 and IL-12 stimulates the production of IFN-γ in Th1 via positive feedback. IFN-γ also inhibits the production of IL-4, an important cytokine that induces the differentiation of naive helper T cells (Th0 cells) to Th2 cells (Ref. Reference Belizário92).

Th2 leads to a humoral immune response by releasing different cytokines (IL-4, IL-5, IL-9, IL-10, IL-13 and IL-25), which stimulate B cells, Th1, eosinophils, basophils or mast cells. Th2 will stimulate B-cell proliferation and antibody production (IgG, IgM, IgA and IgE antibodies). The Th1/Th2 ratio value often correlates with the severity of pancreas injury. The predominant Th2 cells are a marker of favourable evolution and the mediators synthesized by these cells are found in large quantities in MAP. The decrease in the Th1/Th2 ratio correlates with an increased level of IL-6, TNF-α and IFN-γ, which secondary leads to reduced levels of IL-4 (Ref. Reference Rodriguez-Nicolas93). Reversed, in SAP, there is a discrepancy regarding the number of Th1, which persistently increases during the first days of evolution. Furthermore, the ratio of Th1/Th2 in SAP remains altered over time, being elevated even after 336 h (Ref. Reference Zhuang94).

Th9 is a new subgroup of CD4 cells, which secretes IL-9, an important mediator of the enrichment of immune cells, such as eosinophils and mast cells (Ref. Reference Chen95). Even though its role in inflammatory disorders or parasitosis is known, its involvement in AP etiopathogenesis has not yet been described.

Th17 is a unique subset of CD4 cells that plays a significant role in AP by producing IL-17, an important cytokine that protects the body against extracellular bacteria or fungi. After their activation by different cytokines, Th17 expresses IL-21, which is important in RORγt and IL-17 expression, determining T-cell proliferation and counteracting suppression by Tregs. Th17 also produces other inflammatory cytokines (TNF-α, IL-22 and IL-26), which play an important role in inflammation (Refs Reference Acosta-Rodriguez96, Reference Chen97). IL-22 induces antimicrobial proteins, defensins, acute-phase proteins, inflammatory cytokines and chemokines. Many studies have shown that IL-22 protects against AP, reducing acinar cell apoptosis, enhancing acinar cell regeneration and limiting inflammation or fibrosis (Ref. Reference Fu, Xiu and Xu98). IL-26 is a novel proinflammatory cytokine overexpressed in activated or transformed T cells. IL-26 stimulates the production of IL-1β, IL-6 and TNFα by human monocytes and is an essential antimicrobial agent, able to form pores in bacterial membranes (Ref. Reference Larochette99).

Th22 cells are a subpopulation of CD4 T cells implicated in the protective mechanisms against various bacterial and viral pathogens. Through their production of IL-22, these cells are involved in tissue repair processes and regulate innate immune mechanisms by producing chemokines and cytokines (Ref. Reference Sun100). IL-22 has an important antimicrobial role, stimulates innate immunity, maintains tissue integrity and enhances tissue repair. Besides Th22, many other cells produce IL-22 in the body (Th1, Th17 cytotoxic T-cell subsets, γδT cells, NK and NKT cells) (Ref. Reference Wolk101). In SAP, the IL-22 produced by Th22 decreases as the disease progresses. It has also been proven that pulmonary complications in SAP are mediated by melatonin due to the upregulation of IL-22 and Th22 activities (Ref. Reference Huai102). In 2016, Qiao et al. (Ref. Reference Qiao103) described the protective effect of recombinant IL-22 against L-arginine-induced SAP in mice by enhancing the expression of anti-apoptosis genes. Other studies that followed also described the beneficial role of IL-22, especially against the complications of SAP (Ref. Reference Vasseur104). Elevated serum levels of IL-22 have been found in patients with AP, demonstrating the protective role of Th22 by interfering with autophagy processes. Th22 cells can stimulate the production of the anti-apoptotic factors (Bcl-2 and Bcl-XL), binding Beclin-1, an indispensable regulator of autophagy. Thus, Th22 blocks the process of acinar self-destruction, preventing pancreatic injury primarily via IL-22 (Ref. Reference Tran, Fairlie and Lee105).

T follicular helper cells (Tfh) are a specialized subset of CD4+ T cells with a critical role in protective immunity by constantly helping B cells to produce antibodies. Tfh cells are located predominantly in secondary lymphoid organs (tonsils, spleen and lymph nodes) at the level of the B-cell zone, constantly interacting with B cells and favouring their maturation and differentiation (Ref. Reference Mintz and Cyster106). Tfh cells recruited CD8+ T cells and B cells by secreting CXCL13 (chemokine ligand 13) and promoted the maturation of B cells into antibody-producing plasma cells by secreting IL-21. An important number of Tfh are found in the peripheral blood circulation, fulfilling similar roles to germinal ones and releasing many cytokines like (IL-4, IL-6, IL-5, IL-13, IL-17, IL-21 and IL-22) (Ref. Reference Sánchez-Vargas and Mathew107). Tfh cells increased significantly in patients with AP and have a significant role in the development and progression of AP, primarily through the implication of IL-6 and IL-21 (Ref. Reference Sánchez-Vargas and Mathew107).

Treg cells

Tregs are a specialized subpopulation that suppresses other immune cells and prevents the body from attacking itself, maintaining immune homeostasis and self-tolerance in this way. Tregs can inhibit T-cell proliferation and cytokine production, which is essential to protect against autoimmunity (Ref. Reference Kondĕlková108). In AP, Treg activation determines the failure of the duodenal barrier function and favours commensal bacterial translocation into pancreatic necrosis (Ref. Reference Glaubitz109). Tregs also control proinflammatory response by reducing the activity of DC and NK cells, and promoting the differentiation of macrophages in those of type 2, with anti-inflammatory effects. However, in SAP, there is a substantial increase in the percentage of Tregs that is maintained as the pancreas destruction progresses. So, the number of Tregs is an important marker of unfavourable evolution in AP (Ref. Reference Stojanovic86). In AP, Tregs play a significant role in the counterbalance against SIRS syndrome. Infected pancreatic necrosis, a tardive complication of necrotizing AP, is secondary to bacterial translocation from the bowel associated with anti-inflammatory response syndrome (CARS) mediated by Tregs (Ref. Reference Glaubitz109).

Cytotoxic T lymphocytes (CD8 cells)

The role of CD8 cells in the adaptive immune system implies the responses of their T-cell receptors to diverse antigens presented by MHC class I molecules by proliferating, secreting cytokines and chemokines, and directly lysing infected cells (Ref. Reference Berg and Forman110). CD8 cells become activated after they recognize the antigen and start the attack against infected cells in three directions. They secrete cytokines with antimicrobial effects (TNF-α and IFN-γ), produce and release cytotoxic granules (perforin and granzymes) and, probably the most important function, CD8 determines the destruction of infected cells via Fas/FasL interactions (Ref. Reference Reina-Campos, Scharping and Goldrath111). To date, there is no consensus about the implication of CD8 cells in AP. Some authors describe a significant depletion of these cells in SAP, others an increase in the CD8 level, while others reported no difference (Ref. Reference Qiao103, Reference Liu112).

Cytotoxic T lymphocytes and NK cells have complementary roles. If the virus escapes the CD8 action, NK cells will intensify their action. As part of innate immunity, NK cells will recognize infected cells and determine cellular death by secretory lysosome exocytosis (Ref. Reference Topham and Hewitt113).

B cells

B cells play an important immunomodulatory role. They secrete anti-inflammatory cytokines (IL-10, IL-35 and TGF-β) that inhibit the activation and proliferation of proinflammatory immune cells (Ref. Reference Rosser and Mauri114). By producing IL-10, B cells block the immune response of Th1 and Th-17 cells, reducing their proinflammatory activity (Ref. Reference Flores-Borja115). IL-35 is a recently discovered anti-inflammatory cytokine from the IL-12 family produced by a wide range of regulatory lymphocytes. It has an important role in blocking the development of Th1 and Th17 cells (Ref. Reference Behzadi, Behzadi and Ranjbar116). In a study conducted by Aksoy et al. (Ref. Reference Aksoy117), the level of IL-35 was significantly lower in people with AP compared with healthy people, this marker being considered in the diagnosis and follow-up of patients with AP.

In the first stages of AP, there is an increased level of B cells, correlated with mild forms of disease. Reversely, in patients with SAP, a reduced number of circulating B cells, especially of regulatory B cells that produce IL-10, and a decreased level of immunoglobulins M and G (IgM and IgG) are often found. IgG levels seem to be an important prognostic marker, being very low in patients who developed infectious complications or those who died (Ref. Reference Ueda118). Furthermore, increased B-cell-activating factor (BAFF) levels were detected in patients with SAP. BAFF is a major cytokine that regulates B-cell survival, maturation and differentiation. BAFF acts as an acute-phase reactant and, like IL-6 or procalcitonin, can predict the severity of AP; these parameters were found to be significantly higher in patients who developed necrosis or who required intensive care hospitalization for more than 7 days (Ref. Reference Pongratz119).

Studies conducted on mice proved that the value of amylase and the histological score of AP increases inversely proportional to the reduction in B lymphocytes. It is now established that B lymphocytes contribute to the severity of AP but the exact role of B subsets in PA etiopathogenesis still needs to be elucidated.

Biomarkers for prognosis in acute pancreatitis

The first 48 h from onset are critical for identifying patients at risk for severe forms or complications. This timeline is also considered a period in which efficient measures can be taken to prevent severe complications and fatal outcomes (Ref. Reference Silva-Vaz120).

Given that, until now, no marker or scoring system has demonstrated a good predictive value in identifying the severe form, there is a continuous search for new molecules.

Based on the pathophysiological mechanisms involved in the progression of AP, several biomarkers could be helpful in the severity stratification; some already showed promising results and others have not yet been investigated. The most investigated markers have been the proinflammatory cytokines and chemokines like IL-6, IL-8, IL-1, IL-10 and IL-17. These molecules play an important role in the evolution of AP but are also unspecific markers of inflammation. There are fewer studies, but with promising results, regarding the more specific pancreatic markers like TAP, CAPAP and markers of initial cell migration and activation (PMN elastase, ICAM, MMP-9 and MCP-1) (Refs Reference Silva-Vaz120, Reference Meher121).

  • TAP is an amino-acid peptide with a low molecular weight rapidly excreted in urine. It is a cleavage product of trypsinogen, released into the systemic circulation with zymogen granule activation. TAP has been used as an early prognostic marker in AP, being significantly higher in patients with SAP versus MAP in the first hours of evolution (Ref. Reference Frossard122). In 2004, a study from the United Kingdom showed that the level of urinary TAP in patients admitted to hospital within 24 h of the onset of symptoms of AP could predict the disease’s unfavourable evolution. Urinary TAP measuring also proves helpful in the early evaluation of the AP severity (Ref. Reference Johnson123).

  • CAPAP is a peptide released from procarboxypeptidase B and is a marker of the activation of pancreatic enzymes (Ref. Reference Müller124). Both serum and urinary CAPAP can be considered early prognostic factors in predicting the severity of AP (Refs Reference Deng125, Reference Regnér126). Muller et al. (Ref. Reference Müller127) demonstrated that CAPAP levels have a predictive value of over 90% in identifying the development of pancreatic necrosis. CAPAP and TAP are both good prognostic markers, although TAP seems more accurate on the first day of admission (Ref. Reference Saez128).

    These activation peptides of pancreatic proteases (TAP and CAPAP) are important in assessing the severity of AP at the beginning of the disease and they will probably be the basis of new studies to identify those patients who will require intensive care therapy (Ref. Reference Staubli, Oertli and Nebiker129).

  • MCP-1 is a chemokine involved in the attraction of monocytes to the inflammation site (Ref. Reference Liu130). Yang et al. (Ref. Reference Yang131) showed statistically significant differences in the values of MCP-1 at admission between the mild, moderate, and severe forms, making MCP-1 a good marker for risk stratification. Another study that sustained these findings found significant differences in MCP-1 concentrations between controls and AP patients, regardless of the severity (Ref. Reference Cavestro132). A study conducted by Regner et al. (Ref. Reference Regnér133) demonstrated that both blood and urine levels of MCP-1 are higher on the first day of admission and decrease gradually in the following days. Serum MCP-1 levels seem superior in predicting SAP at admission compared to urinary levels (Refs Reference Regnér126, Reference Regnér133).

  • ICAMs (ICAM1) are membrane glycoproteins that promote leukocyte migration to the site of inflammation. ICAM serum values are elevated in AP, especially in the severe or necrotizing forms (Refs Reference Sato, Shibata and Maeda134Reference Perejaslov, Chooklin and Bihalskyy137). Zhu et al. (Ref. Reference Zhu and Jiang138) established a cutoff value of 25 ng/ml for differentiating SAP from MAP with a sensibility and specificity of over 60%. The prognosis accuracy of ICAM was similar to the APACHE II score and superior to IL-6.

  • MMP-9 is an endopeptidase involved in extracellular matrix degradation and, in AP, facilitates neutrophil migration in the pancreas. Neutrophils and macrophages secrete it. Several studies have demonstrated a significant increase in serum values of MMP-9 compared to mild forms or controls. MMP-9 could not discriminate between moderate and severe forms (Refs Reference Chen139Reference Nukarinen141). In another study, MMP-9 showed a very good specificity but low sensibility in predicting SAP at admission (Ref. Reference Nawacki, Grabowska and Głuszek142).

    Vitale et al. (Ref. Reference Vitale143) confirmed these findings in a small study in children with AP, showing significant differences in MMP-9 levels between mild and severe forms, making this a potential marker for paediatric patients, where the need for new markers is even more important.

  • PMN elastase (neutrophil elastase) is an enzyme that results after neutrophil activation and degranulation. In AP, blood levels of elastase are increased. A few studies have shown a correlation between the blood level of elastase and the severity of the disease, proving the ability to serve as an early predicting marker. Domingues-Munos et al. (Ref. Reference Domínguez-Muñoz144) demonstrated a good sensibility and specificity for detecting SAP in the first 48 h. Other studies found the same correlation but with a lower predictive value. PMN (polymorphonuclear neutrophils) elastase seems more accurate in predicting SAP at 48 h of admission than CRP (C-reactive protein) (Refs Reference Robert145Reference Wilson147).

  • TNF-α is a major cytokine involved in the inflammation from AP. TNF-α is secreted from various cells and molecules, from acinar cells to DAMPs, endothelial cells, neutrophils, macrophages and T cells (Ref. Reference Szatmary and Gukovsky148). TNF-α needs enzymatic conversion to its soluble form. Also, the activity is dependent on receptor binding. For these reasons, serum measurements can be difficult and few studies exist (Ref. Reference Szatmary and Gukovsky148). The results regarding the prognosis value of TNF-α are conflicting. One study found no connection between serum levels and severity of AP, whereas another study found higher levels of TNF-α in patients with gallstone AP compared to controls (Refs Reference Exley149, Reference Paajanen150).

  • IL-8 is a chemokine secreted in AP by macrophages, monocytes, activated neutrophils and non-immune cells, like endothelial and epithelial cells, in response to cellular injury (Ref. Reference Makhija and Kingsnorth151). Several studies show the predictive value of IL-8 for severe forms (Refs Reference Silva-Vaz120, Reference Gross152). One study demonstrated a sensibility of 60% and specificity of 80% in predicting progression to SAP (Ref. Reference Rao and Kunte153). Also, IL-8 levels correlate well with infected necrosis and systemic complication development (Ref. Reference Rau154). To date, IL-8 is one of the best predicting markers of outcome but its clinical use is still limited (Ref. Reference Rao and Kunte153).

  • IL-6 is produced by several immune cells alongside injured pancreatic cells and endothelial cells (Ref. Reference Makhija and Kingsnorth151). It is the main cytokine involved in the early phase of inflammation in AP and the most investigated cytokine as a marker for diagnosis and prognosis with promising results. IL-6 has shown good results in severity stratification and good predictive value for developing SAP (Refs Reference Dambrauskas155, Reference Berney156). It also correlates with distant organ failure, especially pulmonary involvement (Ref. Reference Gross152). Compared with other markers or already used scores, IL-6 seems superior in predicting severe forms, with good sensitivity and specificity (Refs Reference Dambrauskas155, Reference Mayer157, Reference Sternby158). However, clinical use is still limited due to rapidly decreased plasma levels and low accessibility (Ref. Reference Makhija and Kingsnorth151).

  • IL-10 is an anti-inflammatory cytokine synthesized by the Th2 cells, B cells and monocytes. IL-10 decreases proinflammatory cytokine levels, especially IL-8 and MCP-1 (Ref. Reference Makhija and Kingsnorth151). There are conflicting results in the prognosis value of IL-6 in AP. Some studies showed increased serum levels in patients with AP compared to controls on the first day of admission. Also, patients with mild AP had higher levels of IL-10 compared to those in severe forms (Refs Reference Pezzilli159, Reference Chen160). On the other hand, a study by Rao et al. (Ref. Reference Rao and Kunte153) found no association between IL-10 and severity.

  • IL-17 is a proinflammatory cytokine secreted by Th17, γδ T cells and NK cells (Ref. Reference Gouda and Bhandary161). Recently, it has become a molecule of interest as a prognosis marker and a therapeutic target in AP (Ref. Reference Li162). Jia et al. (Ref. Reference Jia163) demonstrated that IL-17 is a good marker for severity stratification if measured in the first 48 h of admission. Other studies showed a good predictive value not only for SAP but also for systemic complications and mortality (Refs Reference Vlachos164, Reference Dai, Li and Zhang165). The data available until now demonstrate a good prospective value of IL-17 as a single marker for identifying severe forms, but more studies are needed.

  • IL-22 is a cytokine produced by Th22 and other T cells (Th1 and Th17). Regarding the IL-22 role as a marker of AP severity, opinions differ. Some authors sustained that the IL-22 level is not correlated with the severity of AP, while others demonstrated an important correlation between the IL-22 level and the severity degree of AP (Refs Reference Zhang, Chen and Wang166, Reference Xue, Nguyen and Habtezion167). More studies are needed to understand better the role of this cytokine in the pathogenesis of AP and its predictive value.

  • IL-21 has a broad immunomodulatory spectrum in innate and adaptive immunity, determining immune response suppression due to its inhibitory role on DC activation and promoting the proliferation of the immune cells by IL-10 and B-regulatory cell production (Ref. Reference Zarkavelis, Kefas and Pentheroudakis168). A significant increase of IL-21 (promotor of inflammation) has been found in patients with SAP compared with those who developed medium forms of the disease. Still, the exact role of this cytokine in AP has not been established yet. It is supposed to produce immune paresis by inhibiting Treg cells (Refs Reference Stojanovic86, Reference Thomson169).

  • IL-23 is an important cytokine from the IL-12 family. It has a role in stimulating Th17 cells, neutrophils and macrophages. Il-17/IL-23 plays an essential role in inflammatory diseases (Ref. Reference McGeachy and Cua170). Jia et al. (Ref. Reference Vlachos164) demonstrated significantly increased IL-23 levels in SAP patients compared to that in MAP patients and controls. The same study showed a positive correlation between IL-23, IL-17 and CRP in predicting severe forms.

Conclusions

The pathogenesis of AP has long been investigated, but it still has not been fully understood. It is now clear that the immune system plays a pivotal role in this process. It starts from the local injury to the acinar cell that leads to intracellular changes like calcium overload, zymogen activation and eventually to a form of cellular death, and continues with the activation of innate and adaptive immunity and the systemic involvement of AP. The evolution of the disease is correlated with the degree of immune response and the imbalance between proinflammatory and anti-inflammatory molecules.

Understanding all the mechanisms and molecules involved in the development and progression of AP opens the way not only to possible new therapeutic targets but also for a better prediction of severity.

Based on these findings, new research should aim to find new, efficient and reliable markers for disease severity, as well as molecular targets for treatment.

Acknowledgements

Publication of this paper was supported by the Iuliu Hatieganu University of Medicine and Pharmacy Cluj-Napoca through the institutional Open access programme.

Funding statement

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Competing interest

The authors declare no competing interests.

References

Toh, SK, Phillips, S and Johnson, CD (2000) A prospective audit against national standards of the presentation and management of acute pancreatitis in the South of England. Gut 46(2), 239243.CrossRefGoogle ScholarPubMed
Singla, A et al. (2009) Admission volume determines outcome for patients with acute pancreatitis. Gastroenterology 137(6), 19952001.CrossRefGoogle ScholarPubMed
Peery, AF et al. (2012) Burden of gastrointestinal disease in the United States: 2012 update. Gastroenterology 143(5), 11791187.e3.CrossRefGoogle ScholarPubMed
Sigounas, DE et al. (2011) New prognostic markers for outcome of acute pancreatitis: Overview of reporting in 184 studies. Pancreas 40(4), 522532.CrossRefGoogle ScholarPubMed
Iannuzzi, JP et al. (2022) Global incidence of acute pancreatitis is increasing over time: A systematic review and meta-analysis. Gastroenterology 162(1), 122134.CrossRefGoogle Scholar
Morinville, VD, Barmada, MM and Lowe, ME (2010) Increasing incidence of acute pancreatitis at an American pediatric tertiary care center: Is greater awareness among physicians responsible?. Pancreas 39(1), 58.CrossRefGoogle Scholar
Banks, PA et al. (2013) Classification of acute pancreatitis–2012: Revision of the Atlanta classification and definitions by international consensus. Gut 62(1), 102111.CrossRefGoogle ScholarPubMed
Wang, GJ et al. (2009) Acute pancreatitis: Etiology and common pathogenesis. World Journal of Gastroenterology 15(12), 14271430.CrossRefGoogle ScholarPubMed
Suzuki, M, Sai, JK and Shimizu, T. (2014) Acute pancreatitis in children and adolescents. World Journal of Gastrointestinal Pathophysiology 5(4), 416426.CrossRefGoogle ScholarPubMed
Leung, PS and Ip, SP (2006) Pancreatic acinar cell: Its role in acute pancreatitis. International Journal of Biochemistry and Cell Biology 38(7), 10241030.CrossRefGoogle ScholarPubMed
Logsdon, CD and Ji, B (2013) The role of protein synthesis and digestive enzymes in acinar cell injury. Nature Reviews Gastroenterology & Hepatology 10(6), 362370.CrossRefGoogle ScholarPubMed
Weber, CK and Adler, G (2001) From acinar cell damage to systemic inflammatory response: Current concepts in pancreatitis. Pancreatology 1(4), 356362.CrossRefGoogle ScholarPubMed
Dixit, A et al. (2016) Role of trypsinogen activation in the genesis of pancreatitis. Pancreapedia: Exocrine Pancreas Knowledge Base. https://doi.org/10.3998/panc.2016.25.CrossRefGoogle Scholar
Watanabe, T, Kudo, M and Strober, W (2017) Immunopathogenesis of pancreatitis. Mucosal Immunology 10(2), 283298.CrossRefGoogle ScholarPubMed
Dawra, R et al. (2011) Intra-acinar trypsinogen activation mediates the early stages of pancreatic injury but not inflammation in mice with acute pancreatitis. Gastroenterology 141(6), 22102217.e2.CrossRefGoogle Scholar
Lee, PJ and Papachristou, GI (2019) New insights into acute pancreatitis. Nature Reviews Gastroenterology and Hepatology 16(8), 479496.CrossRefGoogle ScholarPubMed
Halangk, W et al. (2000) Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis. Journal of Clinical Investigation 106(6), 773781.CrossRefGoogle ScholarPubMed
Talukdar, R et al. (2016) Release of cathepsin B in cytosol causes cell death in acute pancreatitis. Gastroenterology 151(4), 747758.e5.CrossRefGoogle ScholarPubMed
Wen, L et al. (2016) Calcium signalling, mitochondria and acute pancreatitis: Avenues for therapy. Pancreapedia: Exocrine Pancreas Knowledge Base. https://doi.org/10.3998/panc.2016.15.CrossRefGoogle Scholar
Voronina, S et al. (2002) Bile acids induce calcium signals in mouse pancreatic acinar cells: Implications for bile-induced pancreatic pathology. Journal of Physiology 540(1), 4955.CrossRefGoogle ScholarPubMed
Pallagi, P et al. (2020) Intracellular Ca2+ signalling in the pathogenesis of acute pancreatitis: Recent advances and translational perspectives. International Journal of Molecular Sciences 21(11), 4005.CrossRefGoogle ScholarPubMed
Peng, S et al. (2018) Galactose protects against cell damage in mouse models of acute pancreatitis. Journal of Clinical Investigation 128(9), 37693778.CrossRefGoogle ScholarPubMed
Raraty, MG et al. (2005) Mechanisms of acinar cell injury in acute pancreatitis. Scandinavian Journal of Surgery 94(2), 8996.CrossRefGoogle ScholarPubMed
Biczo, G et al. (2018) Mitochondrial dysfunction, through impaired autophagy, leads to endoplasmic reticulum stress, deregulated lipid metabolism, and pancreatitis in animal models. Gastroenterology 154(3), 689703.CrossRefGoogle ScholarPubMed
Raraty, M et al. (2000) Calcium-dependent enzyme activation and vacuole formation in the apical granular region of pancreatic acinar cells. Proceedings of the National Academy of Sciences of the United States of America 97(24), 1312613131.CrossRefGoogle ScholarPubMed
Wu, JS et al. (2016) Endoplasmic reticulum stress is activated in acute pancreatitis. Journal of Digestive Disease 17(5), 295303.CrossRefGoogle ScholarPubMed
Grapin-Botton, A (2005) Ductal cells of the pancreas. International Journal of Biochemistry and Cell Biology 37(3), 504510.CrossRefGoogle ScholarPubMed
Hegyi, P et al. (2011) Pancreatic ductal bicarbonate secretion: Challenge of the acinar acid load. Frontiers in Physiology 2, 36.CrossRefGoogle ScholarPubMed
Venglovecz, V, Rakonczay, Z and Hegyi, P (2016) The effects of bile acids on pancreatic ductal cells. Pancreapedia: Exocrine Pancreas Knowledge Base. https://doi.org/10.3998/panc.2016.5.CrossRefGoogle Scholar
Liu, T et al. (2017) NF-κB signalling in inflammation. Signal Transduction and Targeted Therapy 2, 17023.CrossRefGoogle ScholarPubMed
Pai, S and Thomas, R (2008) Immune deficiency or hyperactivity-Nf-kappa illuminates autoimmunity. Journal of Autoimmunity 31, 245251.CrossRefGoogle ScholarPubMed
Jakkampudi, A et al. (2016) NF-κB in acute pancreatitis: Mechanisms and therapeutic potential. Pancreatology 16(4), 477488.CrossRefGoogle ScholarPubMed
O’Brien, J et al. (2018) Overview of microRNA biogenesis, mechanisms of actions, and circulation. Frontiers in Endocrinology (Lausanne) 9, 402.CrossRefGoogle ScholarPubMed
Yang, Y et al. (2020) MicroRNAs in acute pancreatitis: From pathogenesis to novel diagnosis and therapy. Journal of Cell Physiology 235(3), 19481961.CrossRefGoogle ScholarPubMed
Bartel, DP (2009) MicroRNAs: Target recognition and regulatory functions. Cell 136(2), 215233.CrossRefGoogle ScholarPubMed
Qin, T et al. (2014) Expressions of miR-22 and miR-135a in acute pancreatitis. Journal of Huazhong University of Science and Technology – Medical Science 34(2), 225233.CrossRefGoogle ScholarPubMed
Ma, X et al.(2015) The oncogenic microRNA miR-21 promotes regulated necrosis in mice. Nature Communications 6, 7151.CrossRefGoogle ScholarPubMed
Gao, M et al. (2018) Chinese Journal of Cellular and Molecular Immunology 34(10), 931936.Google Scholar
Yu, X, Odenthal, M and Fries, JW (2016) Exosomes as miRNA Carriers: Formation-Function-Future. International Journal of Molecular Sciences 17(12), 2028.CrossRefGoogle ScholarPubMed
Zhang, Y, Yan, L and Han, W (2018) Elevated level of miR-551b-5p is associated with inflammation and disease progression in patients with severe acute pancreatitis. Therapeutic Apheresis and Dialysis 22(6), 649655.CrossRefGoogle ScholarPubMed
Wang, D et al. (2018) MiRNA-155 regulates the Th17/Treg ratio by targeting SOCS1 in severe acute pancreatitis. Frontiers in Physiology 9, 686.CrossRefGoogle ScholarPubMed
Li, X et al. (2018) RNA-Seq analyses of the role of miR-21 in acute pancreatitis. Cellular Physiology and Biochemistry 51(5), 21982211.CrossRefGoogle ScholarPubMed
Yang, B et al. (2014) MicroRNAs in mesenteric lymph and plasma during acute pancreatitis. Annals of Surgery 260(2), 341347.Google Scholar
Kang, R et al. (2014) Cell death and DAMPs in acute pancreatitis. Molecular Medicine 20(1), 466477.CrossRefGoogle ScholarPubMed
Lamkanfi, M and Dixit, VM (2012) Inflammasomes and their roles in health and disease. Annual Review of Cell Developmental Biology 28, 137161.CrossRefGoogle ScholarPubMed
Li, H et al. (2023) New insights into regulatory cell death and acute pancreatitis. Heliyon 9(7), e18036.CrossRefGoogle ScholarPubMed
Al Mamun, A et al. (2022) Pyroptosis in acute pancreatitis and its therapeutic regulation. Apoptosis 27(7–8), 465481.CrossRefGoogle ScholarPubMed
Zhou, J and Sahin-Tóth, M (2011) Chymotrypsin C mutations in chronic pancreatitis. Journal of Gastroenterology and Hepatology 26(8), 12381246.CrossRefGoogle ScholarPubMed
Sarshari, B et al. (2023) The possible role of viral infections in acute pancreatitis: A review of literature. Gastroenterology and Hepatology from Bed to Bench 16(3), 270281.Google ScholarPubMed
Thaker, AM, Mosko, JD and Berzin, TM (2015) Post-endoscopic retrograde cholangiopancreatography pancreatitis. Gastroenterology Report (Oxford) 3(1), 3240.CrossRefGoogle ScholarPubMed
Tiwari, AK et al. (2022) Hypercalcemia – An enigmatic cause of acute pancreatitis. Journal of Clinical and Translational Research 8(3), 176180.Google ScholarPubMed
Grisham, JM, Tran, AH and Ellery, K (2022) Hypertriglyceridemia-induced acute pancreatitis in children: A mini-review. Frontiers in Pediatrics 10, 931336.CrossRefGoogle ScholarPubMed
Murphy, KM, Travers, P and Walport, M Janeway’s Immunobiology, 7th Edn. New York: Garland Science, 2007.Google Scholar
Marshall, J.S. et al. (2018) An introduction to immunology and immunopathology. Allergy, Asthma and Clinical Immunology 14(Suppl 2), 49.CrossRefGoogle ScholarPubMed
Hoque, R (2016) Update on innate immunity and perspectives on metabolite regulation in acute pancreatitis. Current Opinion in Gastroenterology 32(6), 507512.CrossRefGoogle ScholarPubMed
Castanheira, FVS and Kubes, P (2019) Neutrophils and NETs in modulating acute and chronic inflammation. Blood 133(20), 21782185.CrossRefGoogle ScholarPubMed
Peng, C, Li, Z and Yu, X (2021) The role of pancreatic infiltrating innate immune cells in acute pancreatitis. International Journal of Medical Sciences 18(2), 534545.CrossRefGoogle ScholarPubMed
Vonlaufen, A et al. (2007) The role of inflammatory and parenchymal cells in acute pancreatitis. Journal of Pathology 213(3), 239248.CrossRefGoogle ScholarPubMed
Wang, J (2018) Neutrophils in tissue injury and repair. Cell and Tissue Research 371(3), 531539.CrossRefGoogle ScholarPubMed
Habtezion, A and Algul, H (2016) Immune modulation in acute and chronic pancreatitis. Pancreapedia: Exocrine Pancreas Knowledge Base. https://doi.org/10.3998/panc.2016.30CrossRefGoogle Scholar
Wan, J et al. (2021) The role of neutrophils and neutrophil extracellular traps in acute pancreatitis. Frontiers in Cell and Developmental Biology 8, 565758.CrossRefGoogle ScholarPubMed
Zhou, M et al. (2010) Acute lung injury and ARDS in acute pancreatitis: Mechanisms and potential intervention. World Journal of Gastroenterology 16(17), 20942099.CrossRefGoogle ScholarPubMed
Soehnlein, O and Lindbom, L (2010) Phagocyte partnership during the onset and resolution of inflammation. Nature Reviews Immunology 10(6), 427439.CrossRefGoogle ScholarPubMed
Hu, F et al. (2020) Macrophages in pancreatitis: Mechanisms and therapeutic potential. Biomedicine and Pharmacotherapy 131, 110693.CrossRefGoogle ScholarPubMed
Mantovani, A et al. (2004) The chemokine system is in diverse forms of macrophage activation and polarization. Trends in Immunology 25(12), 677686.CrossRefGoogle ScholarPubMed
Mosser, DM and Edwards, JP (2008) Exploring the full spectrum of macrophage activation. Nature Reviews Immunology 8(12), 958969.CrossRefGoogle ScholarPubMed
Mamilos, A et al. (2023) Macrophages: From simple phagocyte to an integrative regulatory cell for inflammation and tissue regeneration – A review of the literature. Cells 12(2), 276.CrossRefGoogle Scholar
Katkar, G and Ghosh, P (2023) Macrophage states: There’s a method in the madness. Trends in Immunology 44(12), 954964.CrossRefGoogle ScholarPubMed
Perides, G et al. (2011) TNF-alpha-dependent regulation of acute pancreatitis severity by Ly-6C(hi) monocytes in mice. Journal of Biological Chemistry 286(15), 1332713335.CrossRefGoogle ScholarPubMed
Samanta, J et al. (2019) Ascites in acute pancreatitis: Not a silent bystander. Pancreatology 19(5), 646652.CrossRefGoogle Scholar
Liu, HB et al. (2006) Role of Kupffer cells in acute hemorrhagic necrotizing pancreatitis-associated lung injury of rats. World Journal of Gastroenterology 12(3), 403407.CrossRefGoogle ScholarPubMed
Gea-Sorlí, S et al. (2011) Activation of lung macrophage subpopulations in experimental acute pancreatitis. Journal of Pathology 223(3), 417424.CrossRefGoogle ScholarPubMed
Gea-Sorlí, S and Closa, D (2010) Role of macrophages in the progression of acute pancreatitis. World Journal of Gastrointestinal Pharmacology and Therapy 1(5), 107111.CrossRefGoogle ScholarPubMed
Xu, D et al. (2020) mTOR-Myc axis drives acinar-to-dendritic cell transition and the CD4+ T cell immune response in acute pancreatitis. Cell Death and Disease 11(6), 416.CrossRefGoogle ScholarPubMed
Bedrosian, AS et al. (2011) Dendritic cells promote pancreatic viability in mice with acute pancreatitis. Gastroenterology 141(5), 19151926.e14.CrossRefGoogle ScholarPubMed
Vivier, E et al. (2008) Functions of natural killer cells. Nature Immunology 9(5), 503510.CrossRefGoogle ScholarPubMed
Zitti, B and Bryceson, YT (2018) Natural killer cells in inflammation and autoimmunity. Cytokine and Growth Factor Reviews 42, 3746.CrossRefGoogle Scholar
Ferrero-Andrés, A et al. (2020) NLRP3 inflammasome-mediated inflammation in acute pancreatitis. International Journal of Molecular Science 21(15), 5386.CrossRefGoogle ScholarPubMed
Martinon, F, Burns, K and Tschopp, J (2002) The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Molecular Cell 10(2), 417426.CrossRefGoogle ScholarPubMed
Tsomidis, I, Voumvouraki, A and Kouroumalis, E (2024) The pathogenesis of pancreatitis and the role of autophagy. Gastroenterology Insight 15(2), 303341.CrossRefGoogle Scholar
Guo, H, Callaway, JB and Ting, JP (2015) Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nature Medicine 21(7), 677687.CrossRefGoogle Scholar
Glaubitz, J et al. (2023) Immune response mechanisms in acute and chronic pancreatitis: Strategies for therapeutic intervention. Frontiers in Immunology 14, 1279539.CrossRefGoogle ScholarPubMed
Hoque, R et al. (2011) TLR9 and the NLRP3 inflammasome link acinar cell death with inflammation in acute pancreatitis. Gastroenterology 141(1), 358369.CrossRefGoogle ScholarPubMed
Algaba-Chueca, F et al. (2017) The expression and activation of the AIM2 inflammasome correlates with inflammation and disease severity in patients with acute pancreatitis. Pancreatology 17(3), 364371.CrossRefGoogle ScholarPubMed
Sendler, M et al. (2020) NLRP3 inflammasome regulates development of systemic inflammatory response and compensatory anti-inflammatory response syndromes in mice with acute pancreatitis. Gastroenterology 158(1), 253269.e14.CrossRefGoogle ScholarPubMed
Stojanovic, B et al. (2023) The emerging roles of the adaptive immune response in acute pancreatitis. Cells 12(11), 1495.CrossRefGoogle ScholarPubMed
Sun, L et al. (2020) Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biology 37, 101759.CrossRefGoogle ScholarPubMed
Zhang, J et al. (1998) Fas-mediated apoptosis and activation-induced T-cell proliferation are defective in mice lacking FADD/Mort1. Nature 392(6673), 296300.CrossRefGoogle ScholarPubMed
Pardalis, V et al. (2014) Expression of Fas (CD95/APO-1) and Fas ligand (FasL) in experimentally-induced acute pancreatitis. Journal of Investigative Surgery 27(2), 6572.CrossRefGoogle Scholar
Xiong, Y et al. (2018) Free total rhubarb anthraquinones protect intestinal injury via regulation of the intestinal immune response in a rat model of severe acute pancreatitis. Frontiers in Pharmacology 9, 75.CrossRefGoogle Scholar
Rangel-Frausto, MS et al. (1995) The natural history of the systemic inflammatory response syndrome (SIRS). A prospective study. JAMA 273(2), 117123.CrossRefGoogle ScholarPubMed
Belizário, JE et al. (2016) Thymic and postthymic regulation of naïve CD4(+) T-cell lineage fates in humans and mice models. Mediators of Inflammation 2016, 9523628.CrossRefGoogle ScholarPubMed
Rodriguez-Nicolas, A et al. (2018) TH1 and TH2 cytokine profiles as predictors of severity in acute pancreatitis. Pancreas 47(4), 400405.CrossRefGoogle ScholarPubMed
Zhuang, Q et al. (2022) Dynamic monitoring of immunoinflammatory response identifies immunoswitching characteristics of severe acute pancreatitis in rats. Frontiers in Immunology 13, 876168.CrossRefGoogle ScholarPubMed
Chen, J et al. (2019) T helper 9 cells: A new player in immune-related diseases. DNA and Cell Biology 38(10), 10401047.CrossRefGoogle Scholar
Acosta-Rodriguez, EV et al. (2007) Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nature Immunology 8(9), 942949.CrossRefGoogle Scholar
Chen, Z et al. (2007) Distinct regulation of interleukin-17 in human T helper lymphocytes. Arthritis and Rheumatology 56(9), 29362946.CrossRefGoogle ScholarPubMed
Fu, X, Xiu, Z and Xu, H (2023) Interleukin-22 and acute pancreatitis: A review. Medicine (Baltimore) 102(44), e35695.CrossRefGoogle ScholarPubMed
Larochette, V et al. (2019) IL-26, a cytokine with roles in extracellular DNA-induced inflammation and microbial defense. Frontiers in Immunology 10, 204.CrossRefGoogle ScholarPubMed
Sun, B T Helper Cell Differentiation and Their Function. Dordrecht, The Netherlands: Springer, 2014.CrossRefGoogle Scholar
Wolk, K et al. (2010) Biology of interleukin-22. Seminars in Immunopathology 32(1), 1731.CrossRefGoogle ScholarPubMed
Huai, JP et al. (2012) Melatonin attenuates acute pancreatitis-associated lung injury in rats by modulating interleukin 22. World Journal of Gastroenterology 18(36), 51225128.CrossRefGoogle ScholarPubMed
Qiao, YY et al. (2016) Interleukin-22 ameliorates acute severe pancreatitis-associated lung injury in mice. World Journal of Gastroenterology 22(21), 50235032.CrossRefGoogle ScholarPubMed
Vasseur, P et al. (2014) High plasma levels of the proinflammatory cytokine IL-22 and the anti-inflammatory cytokines IL-10 and IL-1ra in acute pancreatitis. Pancreatology 14(6), 465469.CrossRefGoogle ScholarPubMed
Tran, S, Fairlie, WD and Lee, EF (2021) BECLIN1: Protein structure, function and regulation. Cells 10(6), 1522.CrossRefGoogle ScholarPubMed
Mintz, MA and Cyster, JG (2020) T follicular helper cells in germinal center B cell selection and lymphomagenesis. Immunological Reviews 296(1), 4861.CrossRefGoogle ScholarPubMed
Sánchez-Vargas, LA and Mathew, A (2019) Peripheral follicular helper T cells in acute viral diseases: A perspective on dengue. Future Virology 14(3), 161169.CrossRefGoogle ScholarPubMed
Kondĕlková, K et al. (2010) Regulatory T cells (TREG) and their roles in immune system with respect to immunopathological disorders. Acta Medica (Hradec Kralove) 53(2), 7377.CrossRefGoogle ScholarPubMed
Glaubitz, J et al. (2023) Activated regulatory T-cells promote duodenal bacterial translocation into necrotic areas in severe acute pancreatitis. Gut 72(7), 13551369.CrossRefGoogle ScholarPubMed
Berg, RE and Forman, J (2006) The role of CD8 T cells in innate immunity and in antigen nonspecific protection. Current Opinion in Immunology 18(3), 338343.CrossRefGoogle Scholar
Reina-Campos, M, Scharping, NE and Goldrath, AW (2021) CD8+ T cell metabolism in infection and cancer. Nature Reviews Immunology 21(11), 718738.CrossRefGoogle ScholarPubMed
Liu, Y et al. (2015) The decrease of peripheral blood CD4+ T cells indicates abdominal compartment syndrome in severe acute pancreatitis. PLoS One 10(8), e0135768.CrossRefGoogle ScholarPubMed
Topham, NJ and Hewitt, EW (2009) Natural killer cell cytotoxicity: How do they pull the trigger? Immunology 128(1), 715.CrossRefGoogle ScholarPubMed
Rosser, EC and Mauri, C (2015) Regulatory B cells: Origin, phenotype, and function. Immunity 42(4), 607612.CrossRefGoogle ScholarPubMed
Flores-Borja, F et al. (2013) CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Science Translational Medicine 5(173), 173ra23.CrossRefGoogle ScholarPubMed
Behzadi, P, Behzadi, E and Ranjbar, R (2016) IL-12 family cytokines: General characteristics, pathogenic microorganisms, receptors, and signalling pathways. Acta Microbiologica et Immunologica Hungarica 63(1), 125.CrossRefGoogle ScholarPubMed
Aksoy, EK et al. (2019) Serum interleukin-35 and pentraxin-3 levels in patients with mild acute pancreatitis. Przeglad Gastroenterologiczny 14(1), 4854.Google ScholarPubMed
Ueda, T et al. (2006) Immunosuppression in patients with severe acute pancreatitis. Journal of Gastroenterology 41(8), 779784.CrossRefGoogle ScholarPubMed
Pongratz, G et al. (2013) B cell activating factor of the tumor necrosis factor family (BAFF) behaves as an acute phase reactant in acute pancreatitis. PLoS One 8(1), e54297.CrossRefGoogle Scholar
Silva-Vaz, P et al. (2020) Multifactorial scores and biomarkers of prognosis of acute pancreatitis: Applications to research and practice. International Journal of Molecular Sciences 21(1), 338.CrossRefGoogle ScholarPubMed
Meher, S et al. (2015) Role of biomarkers in diagnosis and prognostic evaluation of acute pancreatitis. Journal of Biomarkers 2015, 519534.CrossRefGoogle ScholarPubMed
Frossard, JL (2001) Trypsin activation peptide (TAP) in acute pancreatitis: From pathophysiology to clinical usefulness. JOP 2(2), 6977.Google ScholarPubMed
Johnson, CD et al. (2004) Urinary trypsinogen activation peptide as a marker of severe acute pancreatitis. British Journal of Surgery 91(8), 10271033.CrossRefGoogle ScholarPubMed
Müller, CA et al. (2003) A study on the activation peptide released from procarboxypeptidase B (CAPAP) and anionic trypsinogen in patients with acute abdominal disorders of non-pancreatic origin. Pancreatology 3(2), 149155.CrossRefGoogle Scholar
Deng, L et al. (2015) Prediction of the severity of acute pancreatitis on admission by carboxypeptidase-B activation peptide: A systematic review and meta-analysis. Clinical Biochemistry 48(10–11), 740746.CrossRefGoogle ScholarPubMed
Regnér, S et al. (2008) Monocyte chemoattractant protein 1, active carboxypeptidase B and CAPAP at hospital admission are predictive markers for severe acute pancreatitis. Pancreatology 8(1), 4249.CrossRefGoogle ScholarPubMed
Müller, CA et al. (2002) Serum levels of procarboxypeptidase B and its activation peptide in patients with acute pancreatitis and non-pancreatic diseases. Gut 51(2), 229235.CrossRefGoogle ScholarPubMed
Saez, J et al. (2002) Comparative study of the activation peptide of carboxypeptidase B and of trypsinogen as early predictors of the severity of acute pancreatitis. Pancreatology 2, 167187.Google Scholar
Staubli, SM, Oertli, D and Nebiker, CA (2015) Laboratory markers predicting severity of acute pancreatitis. Critical Reviews in Clinical Laboratory Sciences 52(6), 273283.CrossRefGoogle ScholarPubMed
Liu, S et al. (2022) Circulating monocytes in acute pancreatitis. Frontiers in Immunology 13, 1062849.CrossRefGoogle ScholarPubMed
Yang, YZ et al. (2016) Clinical significance of dynamic detection for serum levels of MCP-1, TNF-α and IL-8 in patients with acute pancreatitis. Asian Pacific Journal of Tropical Medicine 9(11), 11111114.CrossRefGoogle ScholarPubMed
Cavestro, GM et al. (2010) Connections between genetics and clinical data: Role of MCP-1, CFTR, and SPINK-1 in the setting of acute, acute recurrent, and chronic pancreatitis. American Journal of Gastroenterology 105(1), 199206.CrossRefGoogle ScholarPubMed
Regnér, S et al. (2008) Protease activation, pancreatic leakage, and inflammation in acute pancreatitis: Differences between mild and severe cases and changes over the first three days. Pancreatology 8(6), 600607.CrossRefGoogle ScholarPubMed
Sato, T, Shibata, W and Maeda, S (2019) Adhesion molecules and pancreatitis. Journal of Gastroenterology 54(2), 99107.CrossRefGoogle ScholarPubMed
Kaufmann, P et al. (1996) Increased plasma concentrations of circulating intercellular adhesion molecule-1 (cICAM-1) in patients with necrotizing pancreatitis. Immunobiology 195(2), 209219.CrossRefGoogle ScholarPubMed
Kaufmann, P et al. (1999) Relation of serial measurements of plasma-soluble intercellular adhesion molecule-1 to severity of acute pancreatitis. American Journal of Gastroenterology 94(9), 24122416.CrossRefGoogle ScholarPubMed
Perejaslov, A, Chooklin, S and Bihalskyy, I (2008) Implication of interleukin 18 and intercellular adhesion molecule (ICAM)-1 in acute pancreatitis. Hepatogastroenterology 55(86–87), 18061813Google ScholarPubMed
Zhu, HH and Jiang, LL (2012) Serum inter-cellular adhesion molecule 1 is an early marker of diagnosis and prediction of severe acute pancreatitis. World Journal of Gastroenterology 18(20), 25542560.CrossRefGoogle Scholar
Chen, P et al. (2006) Serum matrix metalloproteinase 9 as a marker for the assessment of severe acute pancreatitis. Tohoku Journal of Experimental Medicine 208(3), 261266.CrossRefGoogle ScholarPubMed
Guo, J et al. (2012) Serum matrix metalloproteinase-9 is an early marker of pancreatic necrosis in patients with severe acute pancreatitis. Hepatogastroenterology 59(117), 15941598.Google Scholar
Nukarinen, E et al. (2016) Association of matrix metalloproteinases −7, −8 and −9 and TIMP -1 with disease severity in acute pancreatitis. A cohort dtudy. PLoS One 11(8), e0161480.CrossRefGoogle Scholar
Nawacki, L, Grabowska, U and Głuszek, S (2019) Can the matrix metalloproteinases 2 and 9 predict the course of acute pancreatitis in previously healthy patients? Acta Gastroenterologica Belgica 82(4), 501505Google ScholarPubMed
Vitale, DS et al. (2022) Matrix metalloproteinases and their inhibitors in pediatric severe acute pancreatitis. PLoS One 17(2), e0261708.CrossRefGoogle ScholarPubMed
Domínguez-Muñoz, E et al. (2006) Accuracy of plasma levels of polymorphonuclear elastase as early prognostic marker of acute pancreatitis in routine clinical conditions. European Journal of Gastroenterology and Hepatology 18(1), 7983.Google Scholar
Robert, JH et al. (2002) Early prediction of acute pancreatitis: Prospective study comparing computed tomography scans, Ranson, Glascow, Acute Physiology and Chronic Health Evaluation II scores, and various serum markers. World Journal of Surgery 26(5), 612619.CrossRefGoogle ScholarPubMed
Gross, V et al. (1990) Granulocyte elastase in assessment of severity of acute pancreatitis. Comparison with acute-phase proteins C-reactive protein, alpha 1-antitrypsin, and protease inhibitor alpha 2-macroglobulin. Digestive Diseases and Sciences 35(1), 97105.CrossRefGoogle ScholarPubMed
Wilson, RB et al. (2005) Serum elastase in the diagnosis of acute pancreatitis: A prospective study. ANZ Journal of Surgery 75(3), 152156.CrossRefGoogle ScholarPubMed
Szatmary, P and Gukovsky, I (2016) The role of cytokines and inflammation in the genesis of experimental pancreatitis. Pancreapedia: Exocrine Pancreas Knowledge Base. https://doi.org/10.3998/panc.2016.29CrossRefGoogle Scholar
Exley, AR et al. (1992) Endotoxaemia and serum tumour necrosis factor as prognostic markers in severe acute pancreatitis. Gut 33(8), 11261128.CrossRefGoogle ScholarPubMed
Paajanen, H et al. (1995) Serum tumour necrosis factor compared with C-reactive protein in the early assessment of severity of acute pancreatitis. British Journal of Surgery 82(2), 271273.CrossRefGoogle ScholarPubMed
Makhija, R and Kingsnorth, AN (2002) Cytokine storm in acute pancreatitis. Journal of Hepatobiliary Pancreatic Surgery 9(4), 401410.CrossRefGoogle ScholarPubMed
Gross, V et al. (1992) Interleukin-8 and neutrophil activation in acute pancreatitis. European Journal of Clinical Investigation 22(3), 200203.CrossRefGoogle ScholarPubMed
Rao, SA and Kunte, AR (2017) Interleukin-6: An early predictive marker for severity of acute pancreatitis. Indian Journal of Critical Care Medicine 21(7), 424428.CrossRefGoogle Scholar
Rau, B et al. (1997) The potential role of procalcitonin and interleukin 8 in the prediction of infected necrosis in acute pancreatitis. Gut 41(6), 832840.CrossRefGoogle ScholarPubMed
Dambrauskas, Z et al. (2010) Different profiles of cytokine expression during mild and severe acute pancreatitis. World Journal of Gastroenterology 16(15), 18451853.CrossRefGoogle ScholarPubMed
Berney, T et al. (1999) Serum profiles of interleukin-6, interleukin-8, and interleukin-10 in patients with severe and mild acute pancreatitis. Pancreas 18(4), 371377.CrossRefGoogle ScholarPubMed
Mayer, J et al. (2000) Inflammatory mediators in human acute pancreatitis: Clinical and pathophysiological implications. Gut 47(4), 546552.CrossRefGoogle Scholar
Sternby, H et al. (2017) IL-6 and CRP are superior in early differentiation between mild and non-mild acute pancreatitis. Pancreatology 17(4), 550554.CrossRefGoogle ScholarPubMed
Pezzilli, R et al. (1997) Serum interleukin-10 in human acute pancreatitis. Digestive Diseases and Sciences 42(7), 14691472.CrossRefGoogle ScholarPubMed
Chen, CC et al. (1999) Serum interleukin 10 and interleukin 11 in patients with acute pancreatitis. Gut 45(6), 895899.CrossRefGoogle ScholarPubMed
Gouda, MM and Bhandary, YP (2019) Acute lung injury: IL-17a-mediated inflammatory pathway and its regulation by curcumin. Inflammation 42, 1160–9.CrossRefGoogle ScholarPubMed
Li, G et al. (2021) Role of interleukin-17 in acute pancreatitis. Frontiers in Immunology 12, 674803.CrossRefGoogle ScholarPubMed
Jia, R et al. (2015) Increased interleukin-23/17 axis and C-reactive protein are associated with severity of acute pancreatitis in patients. Pancreas 44(2), 321325.CrossRefGoogle ScholarPubMed
Vlachos, S et al. (2014) Serum profiles of M30, M65 and interleukin-17 compared with C-reactive protein in patients with mild and severe acute pancreatitis. Journal of Hepatobiliary Pancreatic Sciences 21(12), 911918.CrossRefGoogle ScholarPubMed
Dai, SR, Li, Z and Zhang, JB (2015) Serum interleukin 17 as an early prognostic biomarker of severe acute pancreatitis receiving continuous blood purification. International Journal of Artificial Organs 38(4) 192198.CrossRefGoogle Scholar
Zhang, JL, Chen, WD and Wang, XY (2010) Expression and significance of IL-22 in serum of patients with acute pancreatitis. Journal of Hepatobiliary Surgery 29 214216.Google Scholar
Xue, J, Nguyen, DT and Habtezion, A (2012) Aryl hydrocarbon receptor regulates pancreatic IL-22 production and protects mice from acute pancreatitis. Gastroenterology 143(6), 16701680.CrossRefGoogle ScholarPubMed
Zarkavelis, G, Kefas, A and Pentheroudakis, G (2017) The emerging role of Interleukin-21 as an antineoplastic immunomodulatory treatment option. Translational Cancer Research 6(S2), 328330.CrossRefGoogle Scholar
Thomson, JE et al, (2019) Transient expression of interleukin-21 in the second hit of acute pancreatitis may potentiate immune paresis in severe acute pancreatitis. Pancreas 48(1), 107112.CrossRefGoogle ScholarPubMed
McGeachy, MJ, Cua, DJ (2007) The link between IL-23 and Th17 cell-mediated immune pathologies. Seminars in Immunology 19(6), 372376.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Multiple cellular mechanisms determine apoptosis/necrosis of the acinar cell. Calcium overload is the central mechanism that activates a cascade of events. The intracellular calcium comes from increased ER release and the extracellular space influx. This calcium overload impaired cellular homeostasis by several mechanisms: activation of the nuclear factor κB (NF-κB) pathway and cytokine synthesis; mitochondrial dysfunction and decreased ATP production followed by cellular death; trypsinogen activation and autodigestion followed by cellular destruction; colocalization of zymogens and lysosomes, ductal cell dysfunction. All these processes determine the destruction of acinar cells through necrosis, apoptosis or pyroptosis.

Figure 1

Table 1. The main mechanisms for the initiation of AP associated with the most important causes

Figure 2

Figure 2. Necrotic acinar cells release proinflammatory cytokines and chemokines. They are crucial in attracting immune cells to the pancreas: neutrophils, macrophages, dendritic cells and natural killer (NK) cells. The neutrophils are the first ones that migrate to the pancreas and determine augmentation of inflammation augmentation through the release of elastase, myeloperoxidase (MPO) and neutrophil extracellular traps (NETs). Proinflammatory cytokines cause activation of resident macrophage as well as migration of blood monocytes. In the pancreas, monocytes differentiate into M1 (proinflammatory secreting macrophages) and M2 (anti-inflammatory secreting macrophages). The M1/M2 ratio imbalance is an important step in the augmentation of inflammation in acute pancreatitis (AP). Nevertheless, the circulating proinflammatory cells activate the resident macrophages from the liver and lung and determine the magnitude of systemic complications. Dendritic cells also migrate in the pancreas tissue and are involved in the secretion of proinflammatory molecules. The NK cells have a bivalent role, either stimulating macrophages and dendritic cell activation or destroying them by cytotoxic effect.

Figure 3

Figure 3. In acute pancreatitis (AP), blood T cells are reduced due to two mechanisms: the overexpression of Fas and FasL ligands on the surface, leading to premature apoptosis and migration to the inflamed pancreas. The T cells differentiate in the Th1, which augments the inflammatory response through proinflammatory cytokine production, and Th2, which secretes anti-inflammatory cytokines. The imbalance between these two types, with the predominance of Th1 response, is one of the most important factors for AP progression. Another important subtype of T cells is Th22, which reduces the inflammation in the pancreas by secreting interleukin 22 (IL-22). The role of B cells is not very well understood, but it seems they can play a bivalent role, pro- and anti-inflammatory.