ArticlePDF AvailableLiterature Review

Acute Pancreatitis: Pathogenesis and Emerging Therapies

Authors:

Abstract and Figures

Acute pancreatitis is a severe inflammatory disorder with limited treatment options. Improved understanding of disease mechanisms has led to new and potential therapies. Here we summarize what we view as some of the most promising new therapies for treating acute pancreatitis, emphasizing the rationale of specific treatments based on disease mechanisms. Targeted pharmacologic interventions are highlighted. We explore potential treatment benefits and risks concerning reducing acute injury, minimizing complications, and improving long-term outcomes. Mechanisms associated with acute pancreatitis initiation, perpetuation, and reconstitution are highlighted, along with potential therapeutic targets and how these relate to new treatments.
Content may be subject to copyright.
Review ARticle
10
Acute pancreatitis: pathogenesis and emerging
therapies
Saif Zamana, Fred Gorelicka,b,c,*
Abstract
Acute pancreatitis is a severe inflammatory disorder with limited treatment options. Improved understanding of disease mecha-
nisms has led to new and potential therapies. Here we summarize what we view as some of the most promising new therapies for
treating acute pancreatitis, emphasizing the rationale of specific treatments based on disease mechanisms. Targeted pharmaco-
logic interventions are highlighted. We explore potential treatment benefits and risks concerning reducing acute injury, minimizing
complications, and improving long-term outcomes. Mechanisms associated with acute pancreatitis initiation, perpetuation, and
reconstitution are highlighted, along with potential therapeutic targets and how these relate to new treatments.
Keywords: Acinar cell, Acute pancreatitis, Calcium signaling, Transporters, Treatment
Introduction
Acute pancreatitis (AP), a sudden inammatory disease of
the pancreas, can range from mild discomfort to a severe,
life-threatening illness. It is one of the most common causes of
gastrointestinal disease-related hospitalizations in the United
States.[1] Past AP annual expenditures in the United States were
estimated to be $2.6 billion.[2] Globally, the pooled incidence of
AP is approximately 34 cases per 100,000 general population,
making it a signicant concern for public health.[3] Moreover,
the global incidence of AP is increasing, with an estimated aver-
age annual percent increase of approximately 3% between 1961
and 2016.[4]
There is considerable morbidity and mortality as well as med-
ical costs associated with AP. Globally, there were approximately
115,000 AP deaths in 2019.[5] In its most severe form, AP can
result in systemic inammatory response syndrome (SIRS), mul-
tiple organ failure (MOF), increased infection rates, and mor-
tality rates of up to 30% in those with severe disease.[6] Though
most AP patients survive the disease, they can experience long-
term consequences, including recurrent AP, chronic pancreatitis,
exocrine and endocrine insufciency, and an increased risk of
pancreatic cancer.[7–9]
Although the risks associated with AP are well known, cur-
rent treatment strategies for AP are supportive and include uid
resuscitation, pain management, and nutritional support.[10]
Current treatment paradigms do not target the underlying
pathophysiological mechanisms of this disease. Investigators
have anticipated that understanding AP’s natural history and
mechanisms will lead to new therapeutic strategies to reduce
this debilitating disease’s short-term and long-term burden;
these goals are just being met. This article aims to delve into
the most foundational mechanisms underlying AP and explore
related new potential treatments that could improve patient
outcomes.
Goals of therapy
The primary goals of AP therapy are multifaceted and aim to
reduce the severity of the acute injury and its complications,
shorten intensive care unit (ICU) and hospital stays, reduce mor-
tality, and decrease the associated medical and quality of life
costs. An unresolved question is whether limiting acute and short-
term injury will reduce the emerging longer-term complications
such as pancreatic exocrine insufciency, diabetes, and pancreatic
cancer. For example, research has highlighted the role of inter-
leukin (IL)-6 as an early predictive marker for severe pancreati-
tis.[11] Other studies have underscored the importance of IL-6 in
driving the progression of preneoplastic pancreatic lesions.[12,13]
These ndings prompt an intriguing question of whether ther-
apies targeting IL-6 in the short-term could potentially reduce
the risk of pancreatic cancer or other long-term complications
in patients with AP and prompt the question of the timing and
length of therapy needed to do so. The potential negative effects
of treatments also need to be considered. For example, what is the
risk of reducing acute inammatory responses to an extent that
might increase the risk of infection or change healing responses?
Thus, the duration of treatment and balance between managing
the acute phase and preventing short-term and long-term compli-
cations need to consider potential off-target effects of treatments.
Mechanisms of pancreatitis injury and recovery vary
over time
Our mechanistic knowledge of pancreatitis comes largely
from in vivo rodent studies and ex vivo studies in rodent and
human pancreatic tissue slices and isolated pancreatic cells.
Conrmation of mechanisms described in rodents in human tis-
sues has been limited. In brief, the initiating event most often
occurs in the pancreatic acinar cell, where multiple forms of
injury can lead to abnormal intracellular calcium signaling.[14]
Though less studied, injury responses also occur in the pancre-
atic duct and endothelial cells early in the disease and contribute
aDepartment of Internal Medicine, Yale School of Medicine, New Haven, CT
06511, bVeteran’s Administration Healthcare System, West Haven, CT 06516,
cDepartment of Cell Biology, Yale School of Medicine, New Haven, CT 06511
Data sharing not applicable to this article as no datasets were generated or
analyzed during the current study.
* Corresponding author: Fred Gorelick, VA Connecticut HealthCare System, Yale
School of Medicine Clinical Campus, 950 Campbell Ave, West Haven, CT 06516.
E-mail: fred.gorelick@yale.edu
Copyright © 2023 The Chinese Medical Association, Published by Wolters
Kluwer Health, Inc. This is an open-access article distributed under the terms of
the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0
(CCBY-NC-ND), where it is permissible to download and share the work provided
it is properly cited. The work cannot be changed in any way or used commercially
without permission from the journal.
Journal of Pancreatology (2024) 7:1
Received: 29 September 2023; Accepted 25 December 2023.
Published online 2 January 2024
http://dx.doi.org/10.1097/JP9.0000000000000168
11
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 www.jpancreatology.com
to disease initiation and perpetuation.[15] Changes in acinar
cell calcium signaling can coordinate pathologic responses in
mitochondria and autophagic pathways,[16] activate digestive
enzymes within the acinar cell, misdirect secretion, and drive
the production of inammatory mediators. A complex-ordered
inammatory cascade follows in the pancreas, likely beginning
with platelets, then neutrophils and inammatory macrophages.
The resolution of AP requires suppressing the acute inamma-
tory cells and activating an anti-inammatory cascade. Other
factors, such as reduced blood ow, vascular injury, vessel occlu-
sion, tissue hypoxia, and neurogenic inammation, can modu-
late the injury’s severity and recovery effectiveness.
Less is known about the factors regulating the AP recov-
ery phase than those mediating early phases of injury or how
the acute therapies discussed below might affect recovery and
long-term AP sequelae. However, in addition to the mechanisms
previously described, one key aspect of tissue recovery is the
clearance of necrotic debris and revascularization.[17] This pro-
cess is mediated by macrophages, which phagocytize necrotic tis-
sue and facilitate recovery.[18] One group has demonstrated that
M1 macrophages dominate during the proinammatory phase
of AP, while M2-like macrophages dominate during pancreas
repair and regeneration. Depletion of M2-like macrophages
during the recovery phase delayed inammation resolution.[19]
The precise mechanisms and signaling pathways governing
the regenerative processes in the pancreas following AP remain
incompletely understood. Furthermore, the long-term conse-
quences of AP, such as the development of new-onset diabetes
and pancreatic insufciency, are areas of growing concern and
active research.[20] In a review of 24 prospective clinical studies
involving 1102 patients with a rst episode of AP, newly diag-
nosed diabetes mellitus developed in 15% of individuals within
12 months after the rst episode of AP.[21] One meta-analysis
of 1795 patients from 39 studies demonstrated that 35% of
patients studied had exocrine pancreatic insufciency after AP
on follow-up after hospital discharge.[9] It remains unclear how
early intervention in AP, particularly strategies aimed at avoid-
ing SIRS, might inuence long-term outcomes such as these.
This has led to initiatives like the Diabetes RElated to Acute
Pancreatitis and its Mechanisms (DREAM) Study, a prospective
cohort study designed to investigate and provide the evidence
needed to screen for, prevent, and treat DM after AP.[22] There
remains a need for continued research into the immediate man-
agement of AP and the long-term monitoring and treatment of
patients to mitigate these secondary complications.
Pathologic responses of the acinar cell that drive AP
and therapeutic targeting
Calcium signaling
There is a consensus that acinar cell calcium signaling changes
usually initiate AP.[14,23] In acinar cells, physiologic cytosolic
calcium signals oscillate, are transient, and are essential for
regulated digestive enzyme secretion. However, in the early
phase of AP, acinar cell cytosolic calcium levels are elevated
above physiologic responses (5–20-fold), prolonged, and
physiologic calcium oscillations disappear. These patholog-
ical, sustained elevations in cytosolic calcium are an early
event in models of AP and are seen in rodent and human aci-
nar cells.[24,25] The changes in cytosolic calcium signaling can
drive mitochondrial dysfunction with the opening of the mito-
chondrial transition pore and subsequent reductions in ATP
levels, intracellular trypsinogen activation,[26] disordered auto-
phagy,[27] endoplasmic reticulum (ER) stress,[28] reduced api-
cal and enhanced basolateral zymogen granule exocytosis,[29]
and tight-junction disruption.[30] Though the direct targets of
this elevated calcium remain unclear and may be manifold, a
role for the calcium-activated phosphatase, calcineurin, in the
pathogenesis of AP has been conrmed by using calcineurin
inhibitors and mice with calcineurin deletions.[31,32] However,
the direct cellular targets of calcineurin that transduce signals
and ultimately mediate acinar pancreatitis responses have yet
to be identied.
Cellular calcium transporters regulate physiologic calcium
homeostasis and can contribute to the early disordered acinar
cell calcium signaling in pancreatitis (Fig. 1). Many transport-
ers are important for calcium homeostasis in pancreatic aci-
nar and are shared by inammatory cells. Intracellularly, the
ER-associated inositol 1,4,5-tris-phosphate receptors (IP3Rs)
and ryanodine receptors (RYRs) enable calcium to move from
the ER stores into the cytosol.[14] Mediators of calcium import
on the plasma membrane include store-operated calcium entry
(SOCE) channels such as the Orai proteins (particularly Orai1)
and transient receptor potential cation (TRPC) channels such
as transient receptor potential cation channel subfamily V 1
(TRPV1) and TRPV6. The discrete homeostatic mechanisms
that regulate calcium entry across the plasma membrane, cal-
cium release from ER stores, and mitochondrial calcium and
lysosomal uxes modulate cytosolic calcium levels under physi-
ologic conditions and during AP (Fig. 1). Lysosomal, mitochon-
drial, and nuclear calcium uxes may modify these responses.
Following a stimulus, acinar cell cytosolic calcium levels are pri-
marily reduced by extrusion across the plasma membrane and
re-uptake into the ER.
Calcium efux across the acinar cell plasma membrane is
critical for lowering cytosolic calcium levels and largely medi-
ated Ca2+ ATPases (PMCA) family of transporters.[33] Na+/Ca2+
exchange has little to no role in acinar cell calcium homeosta-
sis. The primary calcium-regulatory mechanisms are largely
conserved among various cell types, including inammatory
and stellate cells.[23] Knowledge of the regulatory pathways for
calcium entry and extrusion has led to new approaches for AP
treatment. Agents that inhibit Ca2+ entry or enhance Ca2+ extru-
sion are summarized below and, in Figure 2, have shown con-
siderable promise for reducing AP severity.
Figure 1. Schematic representation of cellular Ca2+ transporters and their
role in Ca2+ homeostasis. Multiple transporters and proteins regulate acinar
cell calcium signaling and are relevant to early acute pancreatitis responses.
The schematic illustrates key transporters involved in calcium entry (Orai1,
Piezo, TRPV4), intracellular calcium regulation (RYR, IP3R), and calcium
extrusion (PMCA). These transporters play a crucial role in maintaining cal-
cium balance in pancreatic acinar cells and inflammatory cells, and their dys-
regulation can contribute to early disordered signaling in the pathogenesis
of acute pancreatitis. IP3R = inositol 1,4,5-tris-phosphate receptor, PMCA
= plasma membrane calcium ATPase, RYR = ryanodine receptor, TRPV =
transient receptor potential cation channel subfamily V.
12
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 Journal of Pancreatology
Calcium entry (stromal interaction molecule 1, TRPV,
Piezo)
SOCE responds to decreases in ER calcium by increasing cal-
cium inux through the calcium release-activated calcium
channel Orai1 and its sensor stromal interaction molecule 1
(STIM1), which are located on the plasma membrane and ER,
respectively. When ER calcium is depleted, STIM1 oligomerizes
and translocates to ER-plasma membrane junctions, where it
binds and gates Orai1 to activate SOCE, resulting in calcium
entry into the cell.[34] The prominent role of calcium signaling
in AP’s pathogenesis has focused investigators on regulators of
calcium ion ux as potential AP therapeutic targets. For exam-
ple, CM4620, GSK-7975A, and CM128 are small molecule
inhibitors of Orai1, a component of SOCE channels formed
by STIM1 and Orai complexes that facilitate Ca2+ entry into
pancreatic acinar cells. The Orai1 inhibitors CM4620, GSK-
7975A, and CM128 can signicantly reduce the severity of AP
in rodent models by inhibiting SOCE in pancreatic acinar cells,
conferring substantial therapeutic benets in rodent AP mod-
els.[35,36] This has moved into human trials for AP therapy. In a
phase-2, open-label, dose-response study, 21 patients with AP,
SIRS, and hypoxemia were randomized to receive low-dose or
high-dose CM4620 plus standard of care (SOC) or SOC alone.
CM4620 showed a positive safety prole in this study without
increasing serious adverse events compared to SOC. Patients
treated with CM4620 displayed improved AP severity, better
tolerance to solid foods, reduced persistent SIRS, and decreased
hospitalization durations.[37] These are promising preliminary
ndings.
TRPC channels mediate a signicant portion of the receptor-
stimulated Ca2+ inux. The vanilloid receptor-1 (TRPV1)
was the rst member of the TRPV subgroup within the TRP
family to be discovered, consisting of 6 mammalian members
that act as Ca2+ entry channels responsive to diverse physical
and chemical triggers.[38] Furthermore, mechanical stressors
(trauma, gallstones) can initiate AP responses by stimulating
calcium inux through these channels. The mechanosensitive
ion channel Piezo1 is expressed in pancreatic acinar cells and
plays a key role in pressure-induced pancreatitis. Pressure-
induced activation of the Piezo1 ion channel can trigger the
opening of the TRPV4 channel, resulting in toxic calcium
overload and AP.[39] Elevating pressure in the mouse pancreatic
duct leads to pancreatitis; this can be prevented by blocking
or deleting Piezo1. The Piezo1 antagonist GsMTx4 reduces
the severity of AP in murine models.[40] Additionally, inhibit-
ing Piezo1 function is sufcient to prevent pressure-induced
AP, while activating Piezo1 can induce pancreatitis in normal
mice but not in mice where Piezo1 has been deleted from aci-
nar cells.[40] These ndings suggest that blocking Piezo1 could
prevent pancreatitis caused by trauma, gallstones, or medical
procedures such as endoscopic retrograde cholangiopancrea-
tography (ERCP) that increase pressure in the pancreas. To our
knowledge, there are no clinical trials yet that target Piezo1 or
TRVP channels.
Calcium release from intracellular stores (IP3Rs, RYR)
****RyRs are channel proteins in the ER that facilitate the
release of Ca2+ from ER calcium reservoirs into the cytosol.
Additionally, IP3Rs are channels that regulate the release of Ca2+
from the ER into the cytosol.[41] Our group reported that in rats,
zymogen activation is driven by Ca2+ release regulated by the
RYR.[42] Experimental data demonstrated that blocking RYR or
depleting its Ca2+ pools curtailed zymogen activation without
affecting enzyme secretion.[42] Thus, RYR’s role in mediating
zymogen activation, not enzyme release, underscores its poten-
tial involvement in AP. It was later demonstrated that inhibiting
RYR reduces early pancreatitis injury. Using the RYR inhibi-
tor dantrolene in mice, it was observed that pretreatment sub-
stantially reduced cerulein-induced pancreatitis.[43] Specically,
dantrolene decreased pancreatic trypsin activity and serum amy-
lase levels and improved the overall pancreatic histology and
evidence of cellular damage.[43] Though these ndings suggest
RYR’s activation may have a signicant role in pancreatitis, to
our knowledge, its inhibition has not yet been used prospec-
tively in a clinical setting in the setting of AP. However, dantro-
lene has been used in human clinical trials in phase Ib/IIa trials
of patients with Type 1 Wolfram syndrome, a disorder of ER
calcium homeostasis.[44]
Other small molecules, such as caffeine, have been investi-
gated in the setting of calcium signaling in AP. Caffeine and
its metabolites inhibit pathological IP3R-mediated calcium
signaling in pancreatic acinar cells, a response implicated in
AP initiation. Caffeine reduced the severity of experimentally-
induced AP by cerulein, taurolithocholate acid, and fatty acid
ethyl esters in mice. The protective effects were likely due to
inhibition of IP3R calcium signaling rather than other potential
mechanisms like phosphodiesterase inhibition. However, high
doses of caffeine, up to 25 mg/kg, were required for protective
effects in the animal models, which are not practical for clini-
cal use in humans. The ndings suggest methylxanthine-based
compounds like caffeine might be suitable starting points for
Figure 2. There are multiple potential calcium-regulatory targets for acute pancreatitis therapy. This schematic illustrates the intricate pathways of calcium
homeostasis, highlighting the transporters and key signaling molecules involved. In addition, the schematic displays select drugs that target the indicated Ca2+
regulators, showcasing potential pancreatitis therapies. Many of these calcium-signaling targeted agents have been shown to attenuate acinar cell responses
in acute pancreatitis and reduce AP severity in preclinical rodent models. A few have advanced to clinical trials. AP = acute pancreatitis, IP3R = inositol
1,4,5-tris-phosphate receptor, PMCA = plasma membrane calcium ATPase, TRPV = transient receptor potential cation channel subfamily V.
13
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 www.jpancreatology.com
drug discovery efforts targeting abnormal IP3R calcium signal-
ing as an AP therapy.[45]
Calcium extrusion
Two major mechanisms remove calcium from the cytosol.
A calcium ATPase pumps the ion from the cytosol into the
ER, and plasma membrane calcium extrusion ATPases of the
ATP2B4 family move cytosolic Ca2+ into the extracellular space.
Stimulation of Ca2+ extrusion has strong potential for targeted
AP treatment, as summarized below.
Pancreatic acinar cells and duct cells receive high concen-
trations of islet hormones, most relevantly insulin, through an
islet-to-acinar cell portal system. As seen in a T1DM model and
an insulin-receptor knockout mouse, insulin signaling deciency
sensitizes to AP development. Insulin protects acinar cells in
pancreatitis by providing glycolytically derived ATP to drive the
plasma membrane calcium ATPase (PMCA) pumps that lower
cytoplasmic calcium. However, it may have additional effects on
ER calcium pumps. However, the effective insulin dose for pro-
tecting acinar cells against experimental pancreatitis is higher
than the insulin doses needed to maximally enhance peripheral
glucose uptake in mouse models. Because this would predis-
pose to hypoglycemia and require the use of an insulin clamp
if applied therapeutically, its clinical use for AP therapy is not
practical.[46,47]
Insulin appears to protect acinar cells indirectly by inducing
Akt-mediated phosphorylation of 6-phosphofructo-2-kinase/
fructose-2,6-biphosphatase 2 (PFKFB2), which boosts glycol-
ysis and the ATP generation need to drive calcium export by
a PMCA. This reveals a potential therapeutic strategy to acti-
vate PFKFB2 phosphorylation, eliminating the need for sys-
temic insulin administration.[47] There are several potential
agents in early-phase development, though none have entered
clinical trials.[47] It may be relevant that a recent publication
on insulin therapy for AP suggested that it might reduce the
duration of hospitalization and lower ICU-related disease sever-
ity scores (Acute Physiology and Chronic Health Evaluation II
[APACHE-II]).[48]
Work from Jason Bruce’s group has hypothesized that the
severity of AP might be linked to the progressive loss of insu-
lin secretion, a consequence of collateral damage to pancreatic
β cells.[46] This insulin deciency could subsequently reduce
insulin-driven pancreatic antimicrobial peptide expression
(AMP) and secretion into the gut, leading to gut dysbiosis,
decreased mucous secretion, inammation, bacterial translo-
cation, infected pancreatic necrosis, sepsis, and consequently,
a more severe progression of AP. Although detailed ndings
await publication, the group is exploring a correlative study
between plasma insulin levels and gut AMPs throughout AP.
Such studies aim to discern differences in insulin loss between
severe and mild AP cases, potentially highlighting therapeu-
tic opportunities with insulin mimetics or antimicrobial pep-
tides (Jason I. E. Bruce, PhD, unpublished data, 2019–2022,
Manchester, UK).
The secretory protein renalase, a prosurvival and anti-
inammatory factor, directly activates the PMCA PMCA4b.[49]
PMCA4b stimulation by renalase enhances calcium egress from
the acinar cell cytosol (Gorelick, MD, Desir, MD, unpublished
data, 2013) and also coupled to other cell signals such as AKT
and ERK recombinant human renalase (rRNLS) reduces the
severity of experimental murine pancreatitis in vivo.[50] It also
reduces pancreatitis injury in isolated pancreatic acinar cells
in a PMCA4b-dependent manner.[50] These results demonstrate
the potential of renalase as a therapeutic agent for pancreati-
tis by promoting calcium extrusion and reducing cell injury.
Additionally, it has been demonstrated that amino acids 220-
239 of human renalase1 exhibit the protective effects found
in intact renalase in murine models of acute kidney injury.[51]
Renalase peptides containing human renalase 1 aa220-239 site
are now being investigated for potential therapeutic benets in
preclinical AP models.
Calcium target: calcineurin
In pancreatic acinar cells, increased cytosolic Ca2+ activates cal-
cineurin, a phosphatase that is central to the pathogenesis of
AP.[52,53] Calcineurin inhibitors, such as tacrolimus (FK506) and
cyclosporine A, are often used to manage autoimmune condi-
tions.[54] Past studies have demonstrated that these calcineurin
inhibitors, or the genetic deletion of calcineurin, decrease the
severity of experimental pancreatitis, including in a post-ERCP
model.[55] Targeted deletions of calcineurin suggested distinct
cell-specic roles for the enzyme. Specically, the expression
of calcineurin in hematopoietic cells and neutrophils contrib-
utes to the development of lung inammation associated with
pancreatitis. In contrast, calcineurin’s expression in the pan-
creas contributes to local inammation.[32] As such, the bene-
cial effects observed from blocking or deleting calcineurin
in mitigating pancreatitis depend on the site of its expression.
This observation led to further murine studies investigating the
local effects of calcineurin inhibitors in the pancreatic duct.
Introducing calcineurin inhibitors tacrolimus and cyclosporine
A with radiocontrast into the pancreatic duct in a post-ERCP
AP model dramatically reduced injury without apparent tox-
icity.[55] Additionally, preclinical intraductal delivery of calci-
neurin inhibitor formulations was safe and well tolerated in
mice.[56] This initial work has led to human clinical trials. One
prospective pilot study involved patients who had undergone
ERCP; patients were randomized into a control group and a
group receiving tacrolimus at 8  on the day preceding ERCP
and at 8  on the day of ERCP. Patients randomized to the
tacrolimus group underwent ERCP 2 hours after the morning
tacrolimus dose. All patients were monitored for post-ERCP
pancreatitis using clinical symptoms of worsening abdominal
pain and increased pancreatic enzymes. This trial demonstrated
that oral tacrolimus at a cumulative dose of 4 mg signicantly
decreases the incidence of post-ERCP pancreatitis.[57]
The Rectal INdomethacin, oral TacROlimus, or their com-
bination for the prevention of post-ERCP pancreatitis (INTRO
Trial) is an ongoing randomized, controlled, double-blinded trial
that will assess the role of tacrolimus in post-ERCP pancreati-
tis prophylaxis and pharmacologically optimize this therapeutic
strategy. The trial plans to randomize 4874 patients undergoing
ERCP to receive either 5 mg oral tacrolimus or oral placebo 1 to
2 hours before ERCP. It will follow patient’s post-ERCP for 30
days to assess the incidence of post-ERCP pancreatitis.[58] This
trial has begun and is planned to end in December 2024.
Organelle dysfunction
The initial changes in cytosolic calcium and other cytoplasmic
signaling molecules trigger a coordinated, interrelated series of
pathologic responses in acinar cell organelles. The most criti-
cal involves mitochondria and the autophagic-lysosomal path-
way, each making distinct contributions to acinar cell damage.
Relevant to this review, specic agents may be useful in targeting
individual organelles.
Mitochondrial dysfunction
Mitochondrial dysfunction drives AP through multiple patho-
logic mechanisms, including the generation of reactive oxy-
gen species (ROS) and reactive nitrogen species, reduced ATP
generation, and release of cytochrome C.[59–61] Oxidative stress
damages cell membranes and proteins while activating proin-
ammatory transcription factors like active protein 1 (AP-1)[62]
and nuclear factor kappa-light-chain-enhancer of activated
14
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 Journal of Pancreatology
B cells (NF-kB).[63] Additionally, oxidative stress can damage
mitochondrial proteins and DNA, impairing mitochondrial
function and ultimately causing ATP depletion, which results
in further imbalances in oxidative stress and ion regulation.
Oxidative damage and mitochondrial dysfunction contribute
signicantly to pancreatic acinar cell injury and death in AP.[64]
The mitochondrial permeability transition pore (MPTP) plays
a critical role in the pathogenesis of AP by inducing mitochon-
drial dysfunction and necrosis upon its opening. Its opening
leads to the loss of mitochondrial membrane potential, swelling
of the mitochondria, and necrotic cell death. This process is
particularly relevant in AP. Small molecule inhibitors of cyclo-
philin D, a regulator of the MPTP implicated in AP, have been
investigated in preclinical models. One cyclophilin D inhibi-
tor signicantly protected mitochondrial membrane potential
and reduced necrosis in murine and human pancreatic acinar
cells.[65]
Autophagocytic-lysosomal
In addition to mitochondrial dysfunction, the dysfunction of
other cellular organelles, such as defective autophagy and lyso-
somal function, and dysregulation of vesicular transport and
trafcking pathways can trigger AP. Autophagy is linked to
the activation of pancreatic digestive proteases. Trypsinogen
activation to trypsin is an important early event in the patho-
genesis of AP and occurs along the secretory pathway and
in endolysosomal vacuoles (see “Zymogen activation” sec-
tion).[66–68] Some have proposed that this generation of active
trypsin can cause pancreatic “autodigestion,” which is central
to pancreatitis development.[69] Others, using murine models,
see generation of pancreatic trypsin as less essential to the
pathogenesis of AP.
Experimental and genetic models demonstrate that impaired
autophagy and lysosomal function in pancreatic acinar cells
play a foundational role in the initiation and development of AP.
While mechanistically complex, disruptions at multiple steps,
including defective cathepsin processing, reduced lysosomal
enzyme activities, altered vacuolar H+-ATPase localization,
decreased levels of the lysosomal membrane lysosome-associated
membrane protein (LAMP) proteins, impaired autophagosome
clearance, and blocked lysosomal enzyme targeting, contribute
to dysfunction in this pathway. Regardless of whether dysfunc-
tion occurs during autophagosome formation or completion of
autophagy, the resultant inhibition of autophagic ux leads to
the accumulation of active trypsin in the acinar cell, cytoplasmic
vacuolization, metabolic changes including reduced ATP pro-
duction, and pancreatitis.[70,71]
One group investigated the interaction between premature
intracellular activation of digestive proteases within pancreatic
acini and the systemic inammatory response during severe pan-
creatitis in mice. They found that not only acinar cells but also
inltrating macrophages can activate digestive proteases during
endocytosis of zymogen-containing vesicles, a process depen-
dent on pH and cathepsin B (CTSB), leading to macrophage
activation through NF-κB, ultimately contributing to systemic
inammation and pancreatitis severity.[72] It is possible that tryp-
sinogen in macrophages may account for the generation of pan-
creatic trypsin long after disease onset, with the acinar cell being
its initial source.
Dysfunction in one organelle impacts others through their
metabolic interdependence and signaling crosstalk, ultimately
leading to key features of pancreatitis like intrapancreatic
trypsinogen activation, inammation, and cell death. Targeted
genetic disruption of organelle components like the lysosomal
protein LAMP2 can cause spontaneous pancreatitis in mice,[73]
further demonstrating organelle dysfunction’s key role in dis-
ease pathogenesis.[16] As such, therapeutics to broadly restore
organelle homeostasis might effectively treat pancreatitis.
Inflammatory mediators
Changes in circulating and tissue levels of proinammatory
cytokines (tumor necrosis factor-α [TNF-α], IL-1β, IL-6, IL-8)[74]
and anti-inammatory cytokines (eg, IL-10)[75] during AP have
been well-documented in rodents and shown in human disease.
One such report characterized the trajectories of key proinam-
matory (angiopoietin-2, IL-6, IL-8, monocyte chemoattractant
protein [MCP]-1, resistin) and anti-inammatory (hepatocyte
growth factor [HGF], soluble tumor necrosis factor-alpha recep-
tor 1 [sTNF-αR1]) cytokines over the rst 5 days after onset in
a prospective cohort of AP patients. The study found that proin-
ammatory cytokines were signicantly elevated early in severe
acute pancreatitis (SAP) compared to mild forms but exhibited
a downward trajectory after day 1 in SAP versus milder forms
with at or upward trajectories.[76] Anti-inammatory cytokines
increased over time in mild and severe disease. The ndings indi-
cate that proinammatory cytokine responses occur rapidly and
are time-dependent in SAP. This highlights the need to enroll
SAP subjects early in the disease when investigating immune
mechanisms or designing trials targeting cytokines.
Resistin is a small adipokine implicated in obesity. A
meta-analysis of 11 studies involving 892 AP patients found
that circulating resistin levels were signicantly higher in
patients with SAP than those with mild AP, suggesting that ele-
vated resistin levels may predict AP.[77]
Activin A, a member of the TGF-β superfamily, has recently
emerged as a signicant potential mediator of inammation in
AP. Activin A is released into the bloodstream during inamma-
tory events and modulates inammatory responses.[78] In the set-
ting of AP, serum activin A levels correlate with disease severity
in murine models and patient cohorts,[79] suggesting its potential
as a predictive clinical marker and therapeutic target for severe
cases. Moreover, activin A predictive capability of AP severity
occurs independently of other risk factors such as body mass
index.[80] Follistatin is a protein that specically binds to and
inhibits the actions of activin A.[81] One recent study in murine
models demonstrated that following bacterial lipopolysaccha-
ride administration, activin A rapidly increases in circulation,
predominantly sourced from bone marrow–derived cells, espe-
cially neutrophils, while circulating Follistatin shows a delayed
increase.[82] Additionally, inhibiting activin A with a neutralizing
antibody in mice with AP decreased disease severity.[80] These
results raise the question of whether activin inhibition could be
a viable treatment strategy for AP; to our knowledge, no clinical
studies in human subjects have been done to test the effective-
ness of activin inhibition for AP treatment.
The role of proinammatory cytokines in the pathogenesis of
AP raises the question of whether anti-inammatory molecules
that suppress oxidative stress or inammation reduce AP sever-
ity and its complications.[83] One study found that by suppress-
ing NF-κB proinammatory signaling, resveratrol pretreatment
could attenuate pancreas injury, inammation, and oxidative
stress in a mouse model of hyperlipidemia-induced AP. Whether
this translates to human AP remains to be seen.[84]
As the understanding of cytokine involvement in AP continues
to evolve, the mixed lineage kinase domain-like protein (MLKL)
function emerges as a research focus. MLKL and its activated
form, p-MLKL, were upregulated in the pancreas in a mouse
model of AP, independent of RIPK3, the canonical upstream
activator of MLKL. Knockout of MLKL, but not Ripk3, reduced
the severity of pancreatitis in mice by promoting M2 “healing”
macrophage polarization in the pancreas.[85] This suggests a pro-
tective role for M2 macrophages in AP. This effect was mediated
in part by reducing the release of the chemokine (C-X-C) ligand
(CXCL10) from injured pancreatic acinar cells, which normally
act to polarize macrophages toward the proinammatory M1
phenotype. Neutralization of CXCL10 reduced macrophage
M1 polarization and pancreatitis severity in mice.[85] This sug-
gests inhibition of the MLKL-CXCL10-macrophage axis as
15
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 www.jpancreatology.com
a potential therapeutic approach in AP and highlights a non-
canonical inammation-related role for MLKL separate from its
canonical necroptotic function.
TNF-α inhibition has been demonstrated to reduce injury in
experimental AP. Antibody inhibition of TNF-α reduced dis-
ease severity and improved survival in mouse and rat models
of AP.[86,87] Importantly, delaying TNF-α inhibition until after
pancreatitis onset was more protective than early prophylactic
treatment, suggesting that its role may be time-dependent.[87]
Later studies demonstrated that the TNF-α inhibitor, iniximab,
enhanced the therapeutic effectiveness of octreotide by reducing
biochemical markers of injury, improving organ function, and
reducing pathology scores in a rat model.[88] The results support
the potential of anti–TNF-α therapy as an AP therapy and the
Randomised treatment of Acute Pancreatitis with Iniximab:
Double-blind multi-centre trial (RAPID-I trial), a phase IIb, ran-
domized, double-blind, placebo-controlled, multicenter trial of
iniximab in patients with AP. The RAPID-I is one of the rst
clinical trials in AP that involves a degree of precision medicine.
The trial plans to use transcriptomics to elucidate any genetic
factors in patients that may be linked to improved response to
iniximab.[89]
Recently, lactate has been identied as an inammatory mod-
ulator in acute inammatory pancreatic and liver injury. Hoque
et al[90] demonstrated that lactate, through the lactate receptor
Gi-protein–coupled receptor 81 (GPR81), counteracts the acti-
vation of the NLRP3 inammasome by negatively regulating
Toll-like receptors, leading to reduced inammation and organ
damage. Given its protective effects in mouse models, lactate
and its receptor GPR81 are potential targets of immunomodula-
tory therapy for patients with AP and other acute organ injuries.
This information has provided a mechanistic rationale for clini-
cal studies comparing post-ERCP pancreatitis and AP outcomes
in patients given intravenous lactated Ringer or normal saline
for treating AP. In post-ERCP AP, a clear benet for aggres-
sive hydration with lactated Ringer during ERCP and post-
procedure has been shown.[91] Though small preliminary studies
suggest a therapeutic advantage of intravenous lactated Ringer
over normal saline in hospitalized patients with AP, large pro-
spective clinical studies are planned.
Zymogen activation
A characteristic early AP response is the activation of protease
zymogens, particularly trypsinogen, in the pancreatic acinar cell.
Trypsinogen can also be activated outside the acinar cell after
entering the interstitial space through acinar cell death, baso-
lateral exocytosis, or back diffusion from the pancreatic duct
lumen through disrupted tight junctions, which can become
endocytosed and activated in macrophages. This activation
enhances their inammatory state.[72] Trypsinogen activation in
the acinar cell likely occurs in a non-zymogen granule compart-
ment along the secretory pathway[67,92] and within endocytic ves-
icles.[93] Chvanov et al[94] have demonstrated that large endocytic
vacuoles containing trypsin are formed in experimental models
of AP. These endocytic vacuoles are predicted to rupture and
release trypsin into acinar cell cytosol or exocytosis of trypsino-
gen into the extracellular space.[94] The result is predicted to lead
to inappropriate intracellular protease activity, and trypsin acti-
vation of other digestive proenzymes such as chymotrypsino-
gen, proelastase, procarboxypeptidase, and prophospholipase
A2 could contribute to AP injury.
There is a consensus that lysosomal hydrolases are central to
trypsinogen activation. The “co-localization hypothesis” posits
that in the initial phases of AP, acinar cell digestive zymogens,
such as trypsinogen, actively mix with lysosomal hydrolases
within vesicular compartments in the acinar cell. This merg-
ing is proposed to facilitate trypsinogen activation by the
lysosomal hydrolase CTSB, which can activate trypsinogen in
an acidic environment.[95] Research involving AP mice with a
targeted deletion in the CTSB (gene supports its role). When
subjected to experimental secretagogue-induced pancreatitis,
the CTSB-decient mice exhibited a signicant reduction in
trypsin activity—over 80% less than their wild-type counter-
parts. Correspondingly, the manifestation of pancreatic dam-
age, measured by serum amylase and lipase activities, as well
as the degree of acinar tissue necrosis, was halved in the CTSB-
decient mice.[96] This evidence underpins the role of CTSB in
activating intrapancreatic trypsinogen, thereby supporting the
co-localization hypothesis. However, selective compartmental
acidication may also be important.
Notably, pancreatic protease zymogen activation has not
proven a useful therapeutic target. Whether this reects chal-
lenges to appropriately time the therapy, inability to inhibit
relevant proteases, or lack of importance of intrapancreatic
zymogen activation in mediating AP severity remains unclear.
Pathologic responses of the non-acinar cell that drive
AP and therapeutic targeting
Fat cells and triglycerides
Fat cells and their triglyceride content can modulate AP sever-
ity. Specically, hypertriglyceridemia can cause severe pancre-
atitis. Blood levels of triglycerides directly relate to the risk of
developing AP.[97] Lipases hydrolyze triglycerides to generate
free fatty acids, which can vary in their ability to damage the
pancreas. Unsaturated fatty acids are particularly harmful and
can cause mitochondrial dysfunction, calcium overload, and
the generation of inammatory mediators in pancreatic acinar
cells.[98–100] Additionally, hypertriglyceridemia worsens outcomes
in AP, regardless of the initial cause.[101] This effect appears to
be increased in obese patients and mice, likely due to lipolysis
of intrapancreatic fat.[98] One group found that during AP, pan-
creatic triglyceride lipase (PNLIP) leaks into visceral adipose
tissue, causing excessive lipolysis independent of adipocyte-
autonomous adipose triglyceride lipase, leading to increased non-
esteried fatty acids, more severe organ failure, and reduced
survival. In contrast, this mechanism does not occur in acute
diverticulitis, indicating a specic role of PNLIP-induced lipolysis
in the pathogenesis of organ failure during pancreatitis.[99] These
results suggest that treatments that lower triglycerides or inhibit
relevant lipases could potentially reduce the severity of AP.
Insights into the mechanisms of lipid toxicity are appearing.
In human patients and mouse models, increased free unbound
fatty acids, especially unsaturated fatty acids like linoleic and
oleic acid, can enter and damage immune cells by interacting
with cell membrane phospholipids and mitochondrial mem-
branes. This impairs immune cell functions like phagocyto-
sis, reduces bacterial clearance, and increases susceptibility to
infections during pancreatitis.[102] Additionally, some work has
suggested lipotoxicity from peri-pancreatic fat necrosis is a key
factor in converting mild to severe AP in obesity.[103] These stud-
ies suggest that preventing an increase in unbound fatty acids or
promoting their binding to albumin could reduce infections in
and severity of AP. Inhibition of lipolysis using the lipase inhib-
itor orlistat in an obese mouse model of pancreatitis reduced
pancreatic necrosis, systemic inammation, lung and kidney
injury, hypocalcemia, and mortality.[98] These ndings suggest
that lipotoxicity mediated by UFAs contributes to the severe
outcomes in obese patients with pancreatitis and that treatment
modalities that reduce lipolysis could reduce AP severity.
Pancreatic triacylglycerol lipase and pancreatic lipase-related
protein 2 (PNLIPRP2) may be suitable targets for drug devel-
opment. These 2 proteins are present in fat necrosis in human
and experimental pancreatitis and can efciently hydrolyze tri-
glycerides to toxic unsaturated free fatty acids that cause injury.
In cell models, pancreatic triacylglycerol lipase and PNLIPRP2
16
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 Journal of Pancreatology
caused lipotoxic injury.[104] In mouse models, PNLIP activ-
ity increased during AP, generating excess non-esteried fatty
acids.[99] These ndings suggest pancreatic triacylglycerol lipase
and PNLIPRP2 contribute to local and systemic lipotoxic injury
in severe AP. This further supports pancreatic lipase inhibition
as a potential therapeutic approach in AP and that pancreatic
triacylglycerol lipase and PNLIPRP2 may be suitable drug tar-
gets for this strategy.
Previous studies have examined the potential effects of dietary
factors on AP. Obese individuals who consume more saturated
fat have more saturated visceral fat triglycerides that are resis-
tant to hydrolysis by pancreatic lipase. This reduces the genera-
tion of free fatty acids that can cause lipotoxic injury. In contrast,
in leaner individuals who consume more unsaturated fat, the
more unsaturated visceral triglycerides are readily hydrolyzed
by lipase. This generates high lipotoxic-free fatty acid levels that
worsen inammation and organ failure.[100] The ndings provide
a potential explanation for the “obesity paradox” in AP, where
obesity sometimes seems protective. The results suggest dietary
fat saturation, not just the amount of body fat, contributes to
pancreatitis severity through effects on lipotoxicity.
Although the role of triglycerides in AP pathogenesis
is well established, the impact of the removal of circulat-
ing triglycerides in treating AP remains unclear. In a recent
multicenter cohort study of patients with hypertriglyceridemia-
associated AP (HTG-AP), plasmapheresis, while effective in low-
ering plasma triglycerides, was not linked to reduced incidence
or duration of AP-associated organ failure but was associated
with increased ICU admissions.[105] Other studies have high-
lighted the lack of advantages of apheresis compared to insulin
infusion. In a study comparing continuous insulin infusion and
apheresis in 48 patients, apheresis resulted in a rapid 78.5%
reduction in triglyceride levels after the rst session. In contrast,
insulin infusion led to a 44.4% reduction in the rst 24 hours.
However, despite the effectiveness of apheresis treatments, they
did not offer a distinct advantage over insulin infusion in terms
of prognosis and associated complications for HTG-associated
pancreatitis.[106]
As such, the optimal treatment for lowering triglyceride
levels in patients with HTG-AP is undetermined. There may
be negative consequences, such as ICU admissions, associated
with apheresis and systemic issues, including the lack of wide-
spread availability of apheresis. Larger clinical trials are cur-
rently being conducted to help resolve these questions. The
EarLy Elimination of Fatty Acids iN hypertriglyceridemia-
induced acuTe pancreatitis (ELEFANT) trial is an open-label,
multicenter, adaptive randomized clinical trial that is investi-
gating early elimination of triglycerides and free fatty acids in
hypertriglyceridemia-induced AP in a minimum of 495 patients.
The ELEFANT trial will randomize patients to plasmapheresis,
insulin-heparin treatment, or standard uid therapy within 48
hours of symptom onset. The primary endpoint is a compos-
ite of severe AP or mortality.[107] The results will provide high-
quality evidence on whether early removal of triglycerides and
free fatty acids improves outcomes in hypertriglyceridemia-
induced pancreatitis. This could establish a new treatment
approach targeting the inciting factors in this subset of patients.
Other potential targets
Store-Operated Calcium Entry Associated Regulatory Factor
(SARAF) regulates calcium signaling in pancreatic acinar cells.
In mouse models of AP, SARAF levels initially increase but then
decrease over time, leading to excessive calcium inux into aci-
nar cells and worsening pancreatitis. SARAF knockout mouse
models had more severe pancreatitis, while mice overexpressing
SARAF were protected.[108] This suggests that strategies to stabi-
lize or restore SARAF levels in acinar cells could be a potential
new therapeutic approach for treating AP.
MicroRNAs may serve as a critical target in the treatment
of AP. The microRNA miR-26a is crucial in regulating calcium
signaling and overload in pancreatic acinar cells. MiR-26a levels
are reduced in experimental mouse models and human samples
of AP. Mechanistically, miR-26a directly targets and inhibits
the calcium channels TRPC3 and TRPC6, thereby restricting
pathological calcium elevations and protecting against pancre-
atitis. miR-26a deciency in mice worsened pancreatitis, while
miR-26a overexpression, globally or in acinar cells, markedly
reduced pancreatitis severity. Additionally, administering a miR-
26a mimic mitigated cerulein-induced pancreatitis.[109] This
work highlights miR-26a as an intrinsic checkpoint on acinar
cell calcium overload. It demonstrates its therapeutic potential
to alleviate AP by normalizing pathological calcium signaling,
suggesting the need to investigate further other miRNAs that
may be involved in calcium signaling or other mechanisms
underlying AP development.
Anticoagulants
Heparin and its non-anticoagulant derivatives can protect against
SAP in mouse models. The drugs reduced pancreatic necrosis,
inammation, and macrophage inltration in SAP, independent
of anticoagulant function.[110] Release of high mobility group
box 1 (HMGB-1) from pancreas macrophages, which can drive
inammation and multi-organ damage in SAP, is inhibited by
this drug class and independent of their anticoagulant function.
Reduced HMGB-1 release is associated with reduced intestinal
barrier dysfunction and, lung injury and decreased mortality.[110]
With regard to non-heparin anticoagulants, one group found
that orally administered dabigatran etexilate (with anticoagu-
lant and trypsin-inhibiting activities) could reduce trypsin activ-
ity and had therapeutic efcacy in a cerulein pancreatitis mouse
model (T7K24R), but not in a more aggressive AP model.[111]
This work demonstrates that benzamidine derivatives like dab-
igatran can be potent trypsin inhibitors. However, their efcacy
may be limited by the severity of the pathology and drug con-
centrations in the pancreas.[111] Clinically, a retrospective study
of 190,474 AP patients found that those on anticoagulation
therapy before onset had lower risks of ICU admission, acute
kidney injury, organ failure, and inpatient mortality, suggesting
a therapeutic role in AP.[112] These results suggest that heparin,
non-heparin anticoagulants, and non-anticoagulant heparin
derivatives may be future options for AP treatment.
Hormones
Hormonal regulation plays a critical role in the body’s response
to various forms of stress, including AP. In this context, hor-
mones like ghrelin, leptin, and melatonin are metabolic
regulators and are found to be protective roles in AP. By inter-
acting with immune factors, these hormones may support innate
defense mechanisms that reduce the severity of pancreatitis.
Ghrelin has been shown to exert a protective effect in AP mod-
els by modulating inammatory pathways.[113,114] Specically,
ghrelin decreases the expression of nuclear factor kappa B
(NFκB) and the inammatory signal transduction pathway,[115]
leading to lower levels of inammatory cytokines such as IL-1β
and tumor necrosis factor-α (TNFα).[116] Furthermore, ghrelin’s
protective inuence extends to reducing pancreatitis-associated
lung injury and neutrophil sequestration, showcasing its sys-
temic anti-inammatory potential.[116,117]
When given intraperitoneal or intracerebroventricular,
leptin, another hormone modulating immune response, can
reduce experimental AP severity.[116] The underlying mechanism
involves the engagement of sensory nerves and the neuropeptide
calcitonin gene–related peptide (CGRP), essential for leptin’s
protective action.[118] Leptin enhances pancreatic tissue repair
and decreases lung injuries, similar to ghrelin.[119] Its therapeutic
17
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 www.jpancreatology.com
effects include the activation of the nitric oxide (NO) system,
improvement in pancreatic microcirculation, and the potential
release of glucocorticoids that attenuate inammation.[120,121]
Melatonin, commonly known for regulating circadian
rhythms, also confers protection against acute pancreatic inam-
mation.[114,122] Its administration to animal models before induc-
ing pancreatitis results in a marked reduction of inammation
markers, such as edema and leukocyte inltration. It decreases
proinammatory cytokines while increasing anti-inammatory
IL-10 levels.[114,123] It also diminishes apoptosis and necrosis in
pancreatic tissues and improves pancreatic blood ow, which
aids in the clearance of inammatory mediators.[113,124,125]
These ndings support that ghrelin, leptin, and melatonin
could be integral components of the natural defense system
against pancreatic inammation. Their increased blood levels
during the initial phase of pancreatic inammation represent a
physiological response to suppress or mitigate the inammatory
process within the pancreas, offering a promising avenue for
therapeutic intervention in AP.
An emerging interest in examining the utility of corticoste-
roids for severe AP treatment is reected by a favorable meta-
analysis.[126] The benets of corticosteroids when used in selective
coronavirus disease 2019 (COVID-19) patients may also be a
factor. A 5-year prospective clinical trial based on the Beth Israel
Deaconess Medical Center on corticosteroid use in severe AP is
underway (completion date: 2027; gov ID: NCT05160506) in
the United States.
Gut microbiome
Recent research has underscored the gut microbiome’s inuence
on AP using experimental models.[127] Antibiotic therapy can
improve the course of AP in rodents by reducing innate immune
system activation.[128,129] However, we are unaware of ndings
that show similar benets of antibiotics in clinical AP.
A recent study has also shown how microbial imbalances
may relate to the severity of necrotizing pancreatitis. In a study
comparing the gut bacteria of healthy individuals with AP
patients, marked differences were found in microbial diversity
and composition. Specically, patients with necrotizing pan-
creatitis demonstrated microbial species with altered ketone
body and benzoate metabolism.[130] Enterococcus faecium and
Finegoldia magna were identied as potential biomarkers for
necrotizing pancreatitis and infected necrotizing pancreatitis,
respectively.[130] These ndings suggest that gut microbiota pro-
les could inform early necrotizing pancreatitis diagnosis and
treatment, highlighting the microbiome’s potential as a target
for AP management and intervention. However, probiotics’
role in treating AP remains controversial. A meta-analysis of
13 randomized controlled trials with 950 patients reveals that
supplementing pre-, pro-, and synbiotics to standard enteral
nutrition may reduce hospital stays for severe AP in Chinese
cohorts. However, other clinical outcomes showed no signif-
icant improvement.[131] The Probiotics in Pancreatitis Trial
(PROPATRIA), a multicenter, randomized, double-blind, placebo-
controlled trial of 298 patients with severe AP, demonstrated
that enteral probiotic prophylaxis did not decrease infectious
complications in patients with severe AP and was linked to sig-
nicantly increased mortality and gut ischemia in the probiotic
arm of the trial. This has caused broad caution about their use
in this patient population, though this may change with time
and more information.[132–134] Notably, the authors cautioned
against considering probiotics universally harmless, especially
in critically ill patients.[135] The contrasting results from the prior
studies and debate regarding the results of the PROPATRIA trial
advocate for further large-scale, rigorously designed, and con-
trolled studies to conrm the efcacy and safety of probiotic
supplementation in AP treatment and whether any specic spe-
cies of bacteria may have specic benets in AP patients.
Conclusions
AP remains a disease with signicant morbidity and mortality
despite improvements in supportive care. Pathologic calcium
signaling, mitochondrial dysfunction, organelle stress, zymo-
gen activation, lipotoxicity, and uncontrolled inammation are
key mechanisms that drive acinar cell injury and systemic AP
complications. This review summarizes promising pharmaco-
logic approaches that target these underlying disease processes,
including inhibitors of calcium inux, boosters of calcium
efux, anti-inammatory therapies, antioxidants, and strategies
to lower circulating lipotoxic factors (Table 1).
Table 1
Summary of potential therapeutic targets and agents for AP based on current preclinical studies and early clinical trials
General target Specific target Inhibitor or agonist References
Calcium Orai1 (SOCE) CM4620, GSK-7975A, CM_128 [35, 36]
Calcium TRPV1, TRPV4 [38, 39]
Calcium Piezo1 GsMTx4 [40]
Calcium IP3R Caffeine [45]
Calcium RYR Dantrolene [43, 44]
Calcium PMCA (calcium efflux) Insulin, renalase [47, 49–51]
Calcium Calcineurin Tacrolimus, cyclosporine A [48, 52, 53, 55]
Mitochondria MPTP Cyclophilin D inhibitors [65]
Inflammation TNF-αAnti–TNF-α antibodies, infliximab [86–89]
Inflammation MLKL CXCL10 neutralizing antibody [85]
Inflammation NLRP3 inflammasome Lactate [90, 91]
Triglyceride hydrolysis PNLIP Orlistat [98]
Triglyceride hydrolysis PNLIPRP2 [99]
Triglycerides Circulating Triglycerides Plasmapheresis, insulin [105–107]
SARAF SARAF SARAF stabilizers [108]
MicroRNAs miR-26a miR-26a mimic [109]
HMGB-1 HMGB-1 Heparin derivatives [110]
Trypsin Trypsin Dabigatran [111]
The table lists general target categories, specific molecular targets or pathways within each category, and examples of pharmacological inhibitors, agonists, or other interventions that have shown potential
benefits in experimental models or early human studies of AP.
AP = acute pancreatitis, CXCL = chemokine (C-X-C) ligand, HMGB-1 = high mobility group box 1, IP3R = inositol 1,4,5-tris-phosphate receptor, MLKL = mixed lineage kinase domain-like protein, MPTP
= mitochondrial permeability transition pore, NLRP3 = PMCA = plasma membrane calcium ATPase, PNLIP = pancreatic triacylglycerol lipase, PNLIPRP2 = pancreatic triacylglycerol lipase and pancreatic
lipase-related protein 2, RYR = ryanodine receptor, SARAF = Store-Operated Calcium Entry Associated Regulatory Factor, SOCE = store-operated calcium entry, TNF = tumor necrosis factor, TRPV =
transient receptor potential cation channel subfamily V.
18
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 Journal of Pancreatology
Early clinical trials demonstrate the safety and potential ef-
cacy of interventions like TNF-α inhibition, reducing calcium
entry with Orai1-inhibition, and tacrolimus in reducing the inci-
dence and severity of pancreatitis. Further studies are needed to
show denitively improved clinical outcomes. While additional
basic research is still required to elucidate mechanisms fully,
the translation of novel treatments from preclinical studies to
human trials appears to be accelerating.
Multifunctional therapies that simultaneously address sev-
eral pathologic mechanisms may provide the greatest benet.
Further characterization of pathologic pathways and crosstalk
between organelles and cell types will aid in developing combina-
torial treatments. Improved early diagnosis and risk stratication
will enable therapies to be administered quickly and targeted to
patients most likely to benet. With continued progress in under-
standing disease mechanisms and applying this knowledge to
human trials, the management of AP is poised for major advances
in the coming years. Ultimately, the goal is to move beyond sup-
portive care toward therapeutic interventions interrupting the
underlying disease process and improving short- and long-term
patient outcomes.
Acknowledgments
None.
Author contributions
SZ: Project administration; writing - review & editing. FG:
Project administration; resources; supervision; writing - review
& editing.
Financial support
FG’s research is supported by a Veterans Administration Senior
Clinical Scientist Merit Award (BX003250), a Department of
Defense Investigator Initiated Award (PR220457), and the
Henry and Joan Binder Endowment.
Conflicts of interest
The authors declare no conicts of interest.
Ethics approval
Our review did not involve any clinical or animal experiments
and was analyzed only using published open-source stud-
ies, therefore did not involve the approval of the Institutional
Review Board.
References
[1] Lee PJ, Papachristou GI. New insights into acute pancreatitis. Nat Rev
Gastroenterol Hepatol. 2019;16:479–496.
[2] Peery AF, Dellon ES, Lund J, et al. Burden of gastrointestinal disease
in the United States: 2012 update. Gastroenterology. 2012;143:1179–
1187.e3.
[3] Xiao AY, Tan MLY, Wu LM, et al. Global incidence and mortality
of pancreatic diseases: a systematic review, meta-analysis, and meta-
regression of population-based cohort studies. Lancet Gastroenterol
Hepatol. 2016;1:45–55.
[4] Iannuzzi JP, King JA, Leong JH, et al. Global incidence of acute pan-
creatitis is increasing over time: a systematic review and meta-analysis.
Gastroenterology. 2022;162:122–134.
[5] Li C, Jiang M, Pan C, et al. The global, regional, and national burden
of acute pancreatitis in 204 countries and territories, 1990–2019. BMC
Gastroenterol. 2021;21:332.
[6] Fu C-Y, Yeh C-N, Hsu J-T, et al. Timing of mortality in severe acute
pancreatitis: experience from 643 patients. World J Gastroenterol.
2007;13:1966–1969.
[7] Chatila AT, Bilal M, Guturu P. Evaluation and management of acute
pancreatitis. World J Clin Cases. 2019;7:1006–1020.
[8] Liu J, Wang Y, Yu Y. Meta-analysis reveals an association between
acute pancreatitis and the risk of pancreatic cancer. World J Clin Cases.
2020;8:4416–4430.
[9] Huang W, de la Iglesia-García D, Baston-Rey I, et al. Exocrine pancre-
atic insufciency following acute pancreatitis: systematic review and
meta-analysis. Dig Dis Sci. 2019;64:1985–2005.
[10] de-Madaria E, Buxbaum JL, Maisonneuve P, et al. Aggressive or
moderate uid resuscitation in acute pancreatitis. N Engl J Med.
2022;387:989–1000.
[11] Rao SA, Kunte AR. Interleukin-6: an early predictive marker for sever-
ity of acute pancreatitis. Indian J Crit Care Med. 2017;21:424–428.
[12] van Duijneveldt G, Grifn MDW, Putoczki TL. Emerging roles for
the IL-6 family of cytokines in pancreatic cancer. Clin Sci (Lond).
2020;134:2091–2115.
[13] Del Poggetto E, Ho I-L, Balestrieri C, et al. Epithelial memory of inam-
mation limits tissue damage while promoting pancreatic tumorigenesis.
Science. 2021;373:eabj0486.
[14] Petersen OH, Gerasimenko JV, Gerasimenko OV, et al. The roles of
calcium and ATP in the physiology and pathology of the exocrine pan-
creas. Physiol Rev. 2021;101:1691–1744.
[15] Hegyi P, Rakonczay Z. The role of pancreatic ducts in the pathogenesis
of acute pancreatitis. Pancreatology. 2015;15:S13–S17.
[16] Gukovskaya AS, Gorelick FS, Groblewski GE, et al. Recent insights into
the pathogenic mechanism of pancreatitis: role of acinar cell organelle
disorders. Pancreas. 2019;48:459–470.
[17] Westman J, Grinstein S, Marques PE. Phagocytosis of necrotic debris at
sites of injury and inammation. Front Immunol. 2020;10:3030.
[18] Brouckaert G, Kalai M, Krysko DV, et al. Phagocytosis of necrotic cells
by macrophages is phosphatidylserine dependent and does not induce
inammatory cytokine production. Mol Biol Cell. 2004;15:1089–1100.
[19] Wu J, Zhang L, Shi J, et al. Macrophage phenotypic switch orchestrates
the inammation and repair/regeneration following acute pancreatitis
injury. EBioMedicine. 2020;58:102920.
[20] Winter Gasparoto RCG, Racy MDCJ, De Campos T. Long-term out-
comes after acute necrotizing pancreatitis: what happens to the pan-
creas and to the patient? JOP. 2015;16:159–166.
[21] Das SLM, Singh PP, Phillips ARJ, et al. Newly diagnosed diabetes melli-
tus after acute pancreatitis: a systematic review and meta-analysis. Gut.
2014;63:818–831.
[22] Hart PA, Papachristou GI, Park WG, et al. Rationale and design for
the diabetes related to acute pancreatitis and its mechanisms study: a
prospective cohort study from the type 1 diabetes in acute pancreatitis
consortium. Pancreas. 2022;51:568–574.
[23] Petersen OH. The 2022 George E Palade Medal Lecture: toxic Ca2+
signals in acinar, stellate and endogenous immune cells are important
drivers of acute pancreatitis. Pancreatology. 2023;23:1–8.
[24] Mukherjee R, Mareninova OA, Odinokova IV, et al. Mechanism of
mitochondrial permeability transition pore induction and damage in
the pancreas: inhibition prevents acute pancreatitis by protecting pro-
duction of ATP. Gut. 2016;65:1333–1346.
[25] Lugea A, Waldron RT, Mareninova OA, et al. Human pancreatic acinar
cells: proteomic characterization, physiologic responses, and organellar
disorders in ex vivo pancreatitis. Am J Pathol. 2017;187:2726–2743.
[26] Frick T, Fernandez-del-Castillo C, Bimmler D, et al. Elevated calcium and
activation of trypsinogen in rat pancreatic acini. Gut. 1997;41:339–343.
[27] Sukumaran P, Nascimento Da Conceicao V, Sun Y, et al. Calcium sig-
naling regulates autophagy and apoptosis. Cells. 2021;10:2125.
[28] Groenendyk J, Agellon LB, Michalak M. Calcium signaling and endo-
plasmic reticulum stress. Int Rev Cell Mol Biol. 2021;363:1–20.
[29] Gorelick FS, Thrower E. The acinar cell and early pancreatitis responses.
Clin Gastroenterol Hepatol. 2009;7:S10–S14.
[30] Galli P, Brenna A, De Camilli P, et al. Extracellular calcium and the
organization of tight junctions in pancreatic acinar cells. Exp Cell Res.
1976;99:178–183.
[31] O’Konski MS, Pandol SJ. Effects of caerulein on the apical cytoskeleton
of the pancreatic acinar cell. J Clin Invest. 1990;86:1649–1657.
[32] Wen L, Javed TA, Dobbs AK, et al. The protective effects of calcineurin
on pancreatitis in mice depend on the cellular source. Gastroenterology.
2020;159:1036–1050.e8.
[33] Brini M, Carafoli E. The plasma membrane Ca2+ ATPase and the
plasma membrane sodium calcium exchanger cooperate in the regula-
tion of cell calcium. Cold Spring Harb Perspect Biol. 2011;3:a004168.
[34] Chang C-L, Chen Y-J, Quintanilla CG, et al. EB1 binding restricts
STIM1 translocation to ER–PM junctions and regulates store-operated
Ca2+ entry. J Cell Biol. 2018;217:2047–2058.
19
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 www.jpancreatology.com
[35] Waldron RT, Chen Y, Pham H, et al. The Orai Ca2+ channel inhib-
itor CM4620 targets both parenchymal and immune cells to
reduce inammation in experimental acute pancreatitis. J Physiol.
2019;597:3085–3105.
[36] Wen L, Voronina S, Javed MA, et al. Inhibitors of ORAI1 prevent cyto-
solic calcium-associated injury of human pancreatic acinar cells and
acute pancreatitis in 3 mouse models. Gastroenterology. 2015;149:481–
492.e7.
[37] Bruen C, Miller J, Wilburn J, et al. Auxora for the treatment of patients
with acute pancreatitis and accompanying systemic inammatory
response syndrome: clinical development of a calcium release-activated
calcium channel inhibitor. Pancreas. 2021;50:537–543.
[38] Nilius B, Vriens J, Prenen J, et al. TRPV4 calcium entry chan-
nel: a paradigm for gating diversity. Am J Physiol Cell Physiol.
2004;286:C195–C205.
[39] Sutton R. Parenchymal pressure injury Ca2+ entry mechanism in pan-
creatitis. Cell Calcium. 2020;88:102208.
[40] Romac JM-J, Shahid RA, Swain SM, et al. Piezo1 is a mechanically
activated ion channel and mediates pressure induced pancreatitis. Nat
Commun. 2018;9:1715.
[41] Luciani DS, Gwiazda KS, Yang TB, et al. Roles of IP3R and RyR Ca2+
channels in endoplasmic reticulum stress and beta-cell death. Diabetes.
2009;58:422–432.
[42] Husain SZ, Prasad P, Grant WM, et al. The ryanodine receptor medi-
ates early zymogen activation in pancreatitis. Proc Natl Acad Sci USA.
2005;102:14386–14391.
[43] Orabi AI, Shah AU, Ahmad MU, et al. Dantrolene mitigates caerulein-
induced pancreatitis in vivo in mice. Am J Physiol Gastrointest Liver
Physiol. 2010;299:G196–G204.
[44] Abreu D, Stone SI, Pearson TS, et al. A phase Ib/IIa clinical trial of
dantrolene sodium in patients with Wolfram syndrome. JCI Insight.
2021;6:e145188.
[45] Huang W, Cane MC, Mukherjee R, et al. Caffeine protects against exper-
imental acute pancreatitis by inhibition of inositol 1,4,5-trisphosphate
receptor-mediated Ca 2+ release. Gut. 2017;66:301–313.
[46] Sans MD, Bruce J, Williams JA. Regulation of pancreatic exocrine func-
tion by islet hormones. Pancreapedia Exocrine Pancreas Knowl Base.
2020;1:1–37. doi:10.3998/panc.2020.01.
[47] Bruce JIE, Sánchez-Alvarez R, Sans MD, et al. Insulin protects acinar
cells during pancreatitis by preserving glycolytic ATP supply to calcium
pumps. Nat Commun. 2021;12:4386.
[48] Li J, Chen TR, Gong HL, et al. Intensive insulin therapy in severe acute
pancreatitis: a meta-analysis and systematic review. West Indian Med J.
2012;61:574–579.
[49] Wang L, Velazquez H, Chang J, et al. Identication of a receptor for
extracellular renalase. PLoS One. 2015;10:e0122932.
[50] Kolodecik TR, Reed AM, Date K, et al. The serum protein renalase reduces
injury in experimental pancreatitis. J Biol Chem. 2017;292:21047–21059.
[51] Wang L, Velazquez H, Moeckel G, et al. Renalase prevents AKI indepen-
dent of amine oxidase activity. J Am Soc Nephrol. 2014;25:1226–1235.
[52] Husain SZ, Grant WM, Gorelick FS, et al. Caerulein-induced intracel-
lular pancreatic zymogen activation is dependent on calcineurin. Am J
Physiol Gastrointest Liver Physiol. 2007;292:G1594–G1599.
[53] Muili KA, Ahmad M, Orabi AI, et al. Pharmacological and genetic inhi-
bition of calcineurin protects against carbachol-induced pathological
zymogen activation and acinar cell injury. Am J Physiol Gastrointest
Liver Physiol. 2012;302:G898–G905.
[54] Safarini OA, Keshavamurthy C, Patel P. Calcineurin inhibitors. In:
StatPearls. Orlando, FL: StatPearls Publishing; 2023.
[55] Orabi AI, Wen L, Javed TA, et al. Targeted inhibition of pancreatic aci-
nar cell calcineurin is a novel strategy to prevent post-ERCP pancreati-
tis. Cell Mol Gastroenterol Hepatol. 2017;3:119–128.
[56] Ni J, Khalid A, Lin Y-C, et al. Preclinical safety evaluation of calcineurin
inhibitors delivered through an intraductal route to prevent post-ERCP
pancreatitis demonstrates endocrine and systemic safety. Pancreatology.
2023;23:333–340.
[57] Rao B H, Vincent PK, Nair P, et al. Preventive effect of tacrolimus on
patients with post-endoscopic retrograde cholangiopancreatography
pancreatitis. Clin Endosc. 2022;55:665–673.
[58] Akshintala VS, Husain SZ, Brenner TA, et al. Rectal INdomethacin,
oral TacROlimus, or their combination for the prevention of post-
ERCP pancreatitis (INTRO Trial): protocol for a randomized, con-
trolled, double-blinded trial. Pancreatology. 2022;22:887–893.
[59] Leung PS, Chan YC. Role of oxidative stress in pancreatic inamma-
tion. Antioxid Redox Signal. 2009;11:135–165.
[60] Odinokova IV, Sung K-F, Mareninova OA, et al. Mechanisms regulating
cytochrome c release in pancreatic mitochondria. Gut. 2009;58:431–442.
[61] Biczo G, Vegh ET, Shalbueva N, et al. Mitochondrial dysfunction,
through impaired autophagy, leads to endoplasmic reticulum stress,
deregulated lipid metabolism, and pancreatitis in animal models.
Gastroenterology. 2018;154:689–703.
[62] Ma X, Dong X, Xu Y, et al. Identication of AP-1 as a critical regu-
lator of glutathione peroxidase 4 (GPX4) transcriptional suppression
and acinar cell ferroptosis in acute pancreatitis. Antioxidants (Basel).
2022;12:100.
[63] Robles L, Vaziri ND, Ichii H. Role of oxidative stress in the pathogene-
sis of pancreatitis: effect of antioxidant therapy. Pancreat Disord Ther.
2013;3:112.
[64] Mukherjee R, Criddle DN, Gukovskaya A, et al. Mitochondrial injury
in pancreatitis. Cell Calcium. 2008;44:14–23.
[65] Shore ER, Awais M, Kershaw NM, et al. Small molecule inhibitors of
cyclophilin D to protect mitochondrial function as a potential treat-
ment for acute pancreatitis. J Med Chem. 2016;59:2596–2611.
[66] Sherwood MW, Prior IA, Voronina SG, et al. Activation of trypsinogen
in large endocytic vacuoles of pancreatic acinar cells. Proc Natl Acad
Sci USA. 2007;104:5674–5679.
[67] Otani T, Chepilko SM, Grendell JH, et al. Codistribution of TAP and
the granule membrane protein GRAMP-92 in rat caerulein-induced
pancreatitis. Am J Physiol. 1998;275:G999–G1009.
[68] Krüger B, Albrecht E, Lerch MM. The role of intracellular calcium sig-
naling in premature protease activation and the onset of pancreatitis.
Am J Pathol. 2000;157:43–50.
[69] Voronina S, Chvanov M, De Faveri F, et al. Autophagy, acute pancreati-
tis and the metamorphoses of a trypsinogen-activating organelle. Cells.
2022;11:2514.
[70] Gukovskaya AS, Gukovsky I, Algül H, et al. Autophagy, inamma-
tion, and immune dysfunction in the pathogenesis of pancreatitis.
Gastroenterology. 2017;153:1212–1226.
[71] Mareninova OA, Hermann K, French SW, et al. Impaired auto-
phagic ux mediates acinar cell vacuole formation and trypsino-
gen activation in rodent models of acute pancreatitis. J Clin Invest.
2009;119:3340–3355.
[72] Sendler M, Weiss F-U, Golchert J, et al. Cathepsin B-mediated activa-
tion of trypsinogen in endocytosing macrophages increases severity of
pancreatitis in mice. Gastroenterology. 2018;154:704–718.e10.
[73] Mareninova OA, Sendler M, Malla SR, et al. Lysosome associated mem-
brane proteins maintain pancreatic acinar cell homeostasis: LAMP-2
decient mice develop pancreatitis. Cell Mol Gastroenterol Hepatol.
2015;1:678–694.
[74] Bhatia M, Brady M, Shokuhi S, et al. Inammatory mediators in acute
pancreatitis. J Pathol. 2000;190:117–125.
[75] Pezzilli R, Billi P, Miniero R, et al. Serum interleukin-10 in human acute
pancreatitis. Dig Dis Sci. 1997;42:1469–1472.
[76] Greer PJ, Lee PJ, Paragomi P, et al. Severe acute pancreatitis exhib-
its distinct cytokine signatures and trajectories in humans: a prospec-
tive observational study. Am J Physiol Gastrointest Liver Physiol.
2022;323:G428–G438.
[77] Yang J, Liu M, Wang S, et al. Alteration of peripheral resistin and the
severity of acute pancreatitis: a meta-analysis. Front Med (Lausanne).
2022;9:915152.
[78] Phillips DJ, de Kretser DM, Hedger MP. Activin and related proteins in
inammation: not just interested bystanders. Cytokine Growth Factor
Rev. 2009;20:153–164.
[79] Staudacher JJ, Yazici C, Carroll T, et al. Activin in acute pancreatitis:
potential risk-stratifying marker and novel therapeutic target. Sci Rep.
2017;7:12786.
[80] Thomas AL, Castellanos K, Mancinelli G, et al. Activin A modu-
lates inammation in acute pancreatitis and strongly predicts severe
disease independent of body mass index. Clin Transl Gastroenterol.
2020;11:e00152.
[81] Hemmati-Brivanlou A, Kelly OG, Melton DA. Follistatin, an antagonist
of activin, is expressed in the Spemann organizer and displays direct
neuralizing activity. Cell. 1994;77:283–295.
[82] Wu H, Chen Y, Winnall WR, et al. Acute regulation of activin A and its
binding protein, follistatin, in serum and tissues following lipopolysac-
charide treatment of adult male mice. Am J Physiol Regul Integr Comp
Physiol. 2012;303:R665–R675.
[83] Agah S, Akbari A, Sadeghi E, et al. Resveratrol supplementation and
acute pancreatitis: a comprehensive review. Biomed Pharmacother.
2021;137:111268.
[84] Zhang X, Yang G, Chen Y, et al. Resveratrol pre-treatment alleviated
caerulein-induced acute pancreatitis in high-fat diet-feeding mice via
suppressing the NF-κB proinammatory signaling and improving the
gut microbiota. BMC Complement Med Ther. 2022;22:189.
20
Zaman and Gorelick Journal of Pancreatology (2024) 7:1 Journal of Pancreatology
[85] Peng C, Tu G, Wang J, et al. MLKL signaling regulates macrophage
polarization in acute pancreatitis through CXCL10. Cell Death Dis.
2023;14:155.
[86] Hughes CB, Gaber LW, Mohey el-Din AB, et al. Inhibition of TNF
alpha improves survival in an experimental model of acute pancreati-
tis. Am Surg. 1996;62:8–13.
[87] Norman JG, Fink GW, Messina J, et al. Timing of tumor necrosis fac-
tor antagonism is critical in determining outcome in murine lethal
acute pancreatitis. Surgery. 1996;120:515–521.
[88] Huang Y-X, Li W-D, Jia L, et al. Iniximab enhances the therapeu-
tic effectiveness of octreotide on acute necrotizing pancreatitis in rat
model. Pancreas. 2012;41:849–854.
[89] Garami A, Hegyi P. Precision medicine in pancreatitis: the future of
acute pancreatitis care. Function (Oxf). 2023;4:zqad015.
[90] Hoque R, Farooq A, Ghani A, et al. Lactate reduces liver and pan-
creatic injury in Toll-like receptor- and inammasome-mediated
inammation via GPR81-mediated suppression of innate immunity.
Gastroenterology. 2014;146:1763–1774.
[91] Buxbaum J, Yan A, Yeh K, et al. Aggressive hydration with lactated
Ringer’s solution reduces pancreatitis following endoscopic ret-
rograde cholangiopancreatography. Clin Gastroenterol Hepatol.
2014;12:303–307.e1.
[92] Hofbauer B, Saluja AK, Lerch MM, et al. Intra-acinar cell activation
of trypsinogen during caerulein-induced pancreatitis in rats. Am J
Physiol. 1998;275:G352–G362.
[93] Sendler M, Lerch MM. The complex role of trypsin in pancreatitis.
Gastroenterology. 2020;158:822–826.
[94] Chvanov M, De Faveri F, Moore D, et al. Intracellular rupture,
exocytosis and actin interaction of endocytic vacuoles in pancre-
atic acinar cells: initiating events in acute pancreatitis. J Physiol.
2018;596:2547–2564.
[95] van Acker GJ, Perides G, Steer ML. Co-localization hypothesis: a mecha-
nism for the intrapancreatic activation of digestive enzymes during the early
phases of acute pancreatitis. World J Gastroenterol. 2006;12:1985–1990.
[96] Halangk W, Lerch MM, Brandt-Nedelev B, et al. Role of cathepsin B
in intracellular trypsinogen activation and the onset of acute pancre-
atitis. J Clin Invest. 2000;106:773–781.
[97] Tung Y-C, Hsiao F-C, Lin C-P, et al. High triglyceride variabil-
ity increases the risk of rst attack of acute pancreatitis. Am J
Gastroenterol. 2023;118:1080–1090.
[98] Navina S, Acharya C, DeLany JP, et al. Lipotoxicity causes multisys-
tem organ failure and exacerbates acute pancreatitis in obesity. Sci
Transl Med. 2011;3:107ra110.
[99] De Oliveira C, Khatua B, Noel P, et al. Pancreatic triglyceride lipase medi-
ates lipotoxic systemic inammation. J Clin Invest. 2020;130:1931–1947.
[100] Khatua B, El-Kurdi B, Patel K, et al. Adipose saturation reduces lipo-
toxic systemic inammation and explains the obesity paradox. Sci
Adv. 2021;7:eabd6449.
[101] Kiss L, Fűr G, Pisipati S, et al. Mechanisms linking hypertriglyceri-
demia to acute pancreatitis. Acta Physiol (Oxf). 2023;237:e13916.
[102] Kostenko S, Khatua B, Trivedi S, et al. Amphipathic liponecrosis
impairs bacterial clearance and causes infection during sterile inam-
mation. Gastroenterology. 2023;165:999–1015.
[103] Patel K, Trivedi RN, Durgampudi C, et al. Lipolysis of visceral adipo-
cyte triglyceride by pancreatic lipases converts mild acute pancreatitis
to severe pancreatitis independent of necrosis and inammation. Am J
Pathol. 2015;185:808–819.
[104] Khatua B, Trivedi RN, Noel P, et al. Carboxyl ester lipase may not
mediate lipotoxic injury during severe acute pancreatitis. Am J Pathol.
2019;189:1226–1240.
[105] Cao L, Chen Y, Liu S, et al. Early plasmapheresis among patients
with hypertriglyceridemia-associated acute pancreatitis. JAMA Netw
Open. 2023;6:e2320802.
[106] Araz F, Bakiner OS, Bagir GS, et al. Continuous insulin therapy versus
apheresis in patients with hypertriglyceridemia-associated pancreati-
tis. Eur J Gastroenterol Hepatol. 2022;34:146–152.
[107] Zádori N, Gede N, Antal J, et al. EarLy Elimination of Fatty Acids
iN hypertriglyceridemia-induced acuTe pancreatitis (ELEFANT trial):
protocol of an open-label, multicenter, adaptive randomized clinical
trial. Pancreatology. 2020;20:369–376.
[108] Son A, Ahuja M, Schwartz DM, et al. Ca2+ inux channel inhibi-
tor SARAF protects mice from acute pancreatitis. Gastroenterology.
2019;157:1660–1672.e2.
[109] Du W, Liu G, Shi N, et al. A microRNA checkpoint for Ca2+ signaling
and overload in acute pancreatitis. Mol Ther. 2022;30:1754–1774.
[110] Yang J, Tang X, Wu Q, et al. Heparin protects severe acute pancreatitis
by inhibiting HMGB-1 active secretion from macrophages. Polymers
(Basel). 2022;14:2470.
[111] Pesei ZG, Jancsó Z, Demcsák A, et al. Preclinical testing of dabigatran
in trypsin-dependent pancreatitis. JCI Insight. 2022;7:e161145.
[112] Kröner PT, Wallace MB, Raimondo M, et al. Systemic anticoagulation
is associated with decreased mortality and morbidity in acute pancre-
atitis. Pancreatology. 2021;21:1428–1433.
[113] Jaworek J, Konturek SJ. Hormonal protection in acute pancre-
atitis by ghrelin, leptin and melatonin. World J Gastroenterol.
2014;20:16902–16912.
[114] Jaworek J. Ghrelin and melatonin in the regulation of pancreatic
exocrine secretion and maintaining of integrity. J Physiol Pharmacol.
2006;57:83–96.
[115] Zhou X, Xue C. Ghrelin inhibits the development of acute pancreatitis
and nuclear factor kappaB activation in pancreas and liver. Pancreas.
2009;38:752–757.
[116] Kerem M, Bedirli A, Pasaoglu H, et al. Role of ghrelin and leptin
in predicting the severity of acute pancreatitis. Dig Dis Sci.
2007;52:950–955.
[117] Zhou X, Xue C. Ghrelin attenuates acute pancreatitis-induced
lung injury and inhibits substance P expression. Am J Med Sci.
2010;339:49–54.
[118] Jaworek J, Bonio J, Leja-Szpa A, et al. Sensory nerves in central and
peripheral control of pancreatic integrity by leptin and melatonin. J
Physiol Pharmacol. 2002;53:51–74.
[119] Gultekin FA, Kerem M, Tatlicioglu E, et al. Leptin treatment amelio-
rates acute lung injury in rats with cerulein-induced acute pancreatitis.
World J Gastroenterol. 2007;13:2932–2938.
[120] Warzecha Z, Dembiński A, Ceranowicz P, et al. Inuence of leptin
administration on the course of acute ischemic pancreatitis. J Physiol
Pharmacol. 2002;53:775–790.
[121] Scotece M, Conde J, López V, et al. Adiponectin and leptin: new targets
in inammation. Basic Clin Pharmacol Toxicol. 2014;114:97–102.
[122] Gülben K, Ozdemir H, Berberoğlu U, et al. Melatonin modulates the
severity of taurocholate-induced acute pancreatitis in the rat. Dig Dis
Sci. 2010;55:941–946.
[123] Jaworek J, Konturek SJ, Leja-Szpak A, et al. Role of endogenous mel-
atonin and its MT2 receptor in the modulation of caerulein-induced
pancreatitis in the rat. J Physiol Pharmacol. 2002;53:791–804.
[124] Baykal A, Iskit AB, Hamaloglu E, et al. Melatonin modulates mesen-
teric blood ow and TNFalpha concentrations after lipopolysaccha-
ride challenge. Eur J Surg. 2000;166:722–727.
[125] Muñoz-Casares FC, Padillo FJ, Briceño J, et al. Melatonin reduces
apoptosis and necrosis induced by ischemia/reperfusion injury of the
pancreas. J Pineal Res. 2006;40:195–203.
[126] Dong L-H, Liu Z-M, Wang S-J, et al. Corticosteroid therapy for severe
acute pancreatitis: a meta-analysis of randomized, controlled trials.
Int J Clin Exp Pathol. 2015;8:7654–7660.
[127] Patel BK, Patel KH, Bhatia M, et al. Gut microbiome in acute pan-
creatitis: a review based on current literature. World J Gastroenterol.
2021;27:5019–5036.
[128] Jia L, Chen H, Yang J, et al. Combinatory antibiotic treatment pro-
tects against experimental acute pancreatitis by suppressing gut bac-
terial translocation to pancreas and inhibiting NLRP3 inammasome
pathway. Innate Immun. 2020;26:48–61.
[129] Wu L, Hu J, Yi X, et al. Gut microbiota interacts with inamma-
tory responses in acute pancreatitis. Therap Adv Gastroenterol.
2023;16:17562848231202133.
[130] Zou M, Yang Z, Fan Y, et al. Gut microbiota on admission as pre-
dictive biomarker for acute necrotizing pancreatitis. Front Immunol.
2022;13:988326.
[131] Tian X, Pi Y-P, Liu X-L, et al. Supplemented use of pre-, pro-, and syn-
biotics in severe acute pancreatitis: an updated systematic review and
meta-analysis of 13 randomized controlled trials. Front Pharmacol.
2018;9:690.
[132] Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic pro-
phylaxis in predicted severe acute pancreatitis: a randomised,
double-blind, placebo-controlled trial. Lancet. 2008;371:651–659.
[133] Reddy BS, MacFie J. Probiotic prophylaxis in predicted severe acute
pancreatitis. Lancet. 2008;372:113.
[134] Reid G, Gibson G, Sanders ME, et al. Probiotic prophylaxis in pre-
dicted severe acute pancreatitis. Lancet. 2008;372:112–113.
[135] Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic prophy-
laxis in predicted severe acute pancreatitis—authors’ reply. Lancet.
2008;372:114.
How to cite this article: Zaman S, Gorelick F. Acute pancreatitis: pathogenesis
and emerging therapies. J Pancreatol 2024;7:10–20. doi: 10.1097/
JP9.0000000000000168
... Despite the enhanced understanding of AP pathogenesis, no specific therapies are currently approved by the Food and Drug Administration for its treatment (Zaman and Gorelick 2024;Trikudanathan et al. 2024). Management of this disease is limited to supportive care with fluid resuscitation, pain control, and nutrition, reinforcing the need to develop new therapeutic compounds (Sun et al. 2024). ...
Article
Full-text available
Acute pancreatitis is a potentially life-threatening inflammatory disorder of the exocrine pancreas characterized by early activation of pancreatic enzymes followed by macrophage-driven inflammation, and pancreatic acinar cell death. The most common causes are gallstones and excessive alcohol consumption. Inflammation and oxidative stress play critical roles in its pathogenesis. Despite increasing incidence, currently, no specific drug therapy is available to treat or prevent acute pancreatitis, in particular severe acute pancreatitis. New therapeutic agents are very much needed. Plant polyphenols have attracted extensive attention in the field of acute pancreatitis due to their diverse pharmacological properties. In this review, we discuss the potential of plant polyphenols in inhibiting the occurrence and development of acute pancreatitis via modulation of inflammation, oxidative stress, calcium overload, autophagy, and apoptosis, based on the currently available in vitro, in vivo animal and very few clinical human studies. We also outline the opportunities and challenges in the clinical translation of plant polyphenols for the treatment of the disease. We concluded that plant polyphenols have a potential therapeutic effect in the management and treatment of acute pancreatitis. Knowledge gained from this review will hopefully inspire new research ideas and directions for the development and application of plant polyphenols for treating this disease.
... This process is primarily driven by the premature activation of trypsinogen into trypsin within acinar cells. In addition, early injury responses in the pancreatic duct and endothelial cells further contribute to the initiation and perpetuation of the disease (5). The premature activation leads to tissue injury and the release of damage-associated molecular patterns (DAMPs), which triggers neutrophil recruitment and a subsequent inflammatory cascade (6)(7)(8) . ...
Preprint
Full-text available
Acute pancreatitis (AP) is a complex condition requiring immediate treatment. Both extracellular vesicles derived from human umbilical cord mesenchymal stem cells (hUC-MSC-EVs) and emodin, a naturally occurring anthraquinone used in traditional Chinese medicine, have shown therapeutic potential in treating AP. However, the mechanisms by which hUC-MSC-EVs and emodin alleviate AP, and whether they exert a synergistic effect on inflamed pancreatic tissues, remain unclear. In this study, we developed AP cell, organoid, and animal models to compare the effects of emodin, hUC-MSC-EVs, and emodin-loaded hUC-MSC-EVs on cell viability, inflammation, and pyroptosis. Our data revealed that all three treatments improved cell viability, reduced pro-inflammatory cytokine expression, and inhibited pyroptosis in the AP models. Notably, the encapsulation of emodin significantly enhanced the protective effects of hUC-MSC-EVs. These findings suggest that emodin’s protective effects on inflamed pancreatic tissues may be attributed, at least in part, to its anti-inflammatory and anti-pyroptotic properties. Additionally, our study proposes a novel strategy for engineering hUC-MSC-EVs for potential therapeutic applications in AP treatment.
... Contributing factors include genetic predisposition, gallstones, and alcohol misuse (Gukovskaya et al., 2017). The prevailing understanding posits acinar cell injury as the initial trigger, leading to parenchymal necrosis and inflammation, central to the disease's pathology Zaman and Gorelick, 2024). In addition, certain vacuole-associated proteins, including LC3 protein, lysosomal-associated membrane protein (LAMP)-1, and LAMP-2 in autophagic vacuoles, have been revealed to contribute greatly to the modulation of pancreatitis Mareninova et al., 2020). ...
Article
Full-text available
Pancreatic diseases such as pancreatitis and pancreatic cancer represent significant health challenges characterized by high mortality rates and limited survival durations. Autophagy, a crucial cellular catabolic process, has emerged as a focal point in understanding various pathological conditions, spanning inflammation-related disorders to malignant neoplasms. This comprehensive review aims to elucidate the biological intricacies of autophagy and its pivotal roles within two extensively researched pancreatic diseases, namely pancreatitis and pancreatic cancer, drawing upon recent scholarly contributions. The discussion will delve into the nuanced mechanisms underlying autophagy’s involvement in these diseases, shedding light on its potential as a therapeutic target. Furthermore, the review will explore cutting-edge therapeutic interventions leveraging autophagy regulation for managing pancreatitis and pancreatic cancer. Through this analysis, we endeavor to offer novel insights into the pathophysiology of pancreatic disorders and contribute to the development of innovative therapeutic modalities in this challenging clinical domain.
... Once the CL is oxidized, MPTP is opened in a synergic action of Ca 2+ ions, increasing the permeability of the mitochondrial inner membrane and leading to cell death [125,126]. Mitochondrial dysfunction impairs ATP-dependent mechanisms to reduce cytosolic Ca 2+ , and then pathological Ca 2+ elevation triggers other cytotoxic pathways such as premature trypsinogen activation, autophagy impairment and activation of the NF-κB pathway to induce AP [89,127,128]. ...
Article
Full-text available
Acute pancreatitis (AP) is one of the most common gastrointestinal tract diseases with significant morbidity and mortality. Current treatments remain unspecific and supportive due to the severity and clinical course of AP, which can fluctuate rapidly and unpredictably. Mitochondria, cellular power plant to produce energy, are involved in a variety of physiological or pathological activities in human body. There is a growing evidence indicating that mitochondria damage-associated molecular patterns (mtDAMPs) play an important role in pathogenesis and progression of AP. With the pro-inflammatory properties, released mtDAMPs may damage pancreatic cells by binding with receptors, activating downstream molecules and releasing inflammatory factors. This review focuses on the possible interaction between AP and mtDAMPs, which include cytochrome c (Cyt c), mitochondrial transcription factor A (TFAM), mitochondrial DNA (mtDNA), cardiolipin (CL), adenosine triphosphate (ATP) and succinate, with focus on experimental research and potential therapeutic targets in clinical practice. Preventing or diminishing the release of mtDAMPs or targeting the mtDAMPs receptors might have a role in AP progression.
Article
Full-text available
The pathogenesis of acute and chronic pancreatitis has recently evolved as new findings demonstrate a complex mechanism operating through various pathways. In this review, the current evidence indicating that several mechanisms act in concert to induce and perpetuate pancreatitis were presented. As autophagy is now considered a fundamental mechanism in the pathophysiology of both acute and chronic pancreatitis, the fundamentals of the autophagy pathway were discussed to allow for a better understanding of the pathophysiological mechanisms of pancreatitis. The various aspects of pathogenesis, including trypsinogen activation, ER stress and mitochondrial dysfunction, the implications of inflammation, and macrophage involvement in innate immunity, as well as the significance of pancreatic stellate cells in the development of fibrosis, were also analyzed. Recent findings on exosomes and the miRNA regulatory role were also presented. Finally, the role of autophagy in the protection and aggravation of pancreatitis and possible therapeutic implications were reviewed.
Article
Full-text available
Acute pancreatitis (AP) is one of the most common acute abdominal conditions, and its incidence has been increasing for years. Approximately 15–20% of patients develop severe AP (SAP), which is complicated by critical inflammatory injury and intestinal dysfunction. AP-associated inflammation can lead to the gut barrier and function damage, causing dysbacteriosis and facilitating intestinal microbiota migration. Pancreatic exocrine deficiency and decreased levels of antimicrobial peptides in AP can also lead to abnormal growth of intestinal bacteria. Meanwhile, intestinal microbiota migration influences the pancreatic microenvironment and affects the severity of AP, which, in turn, exacerbates the systemic inflammatory response. Thus, the interaction between the gut microbiota (GM) and the inflammatory response may be a key pathogenic feature of SAP. Treating either of these factors or breaking their interaction may offer some benefits for SAP treatment. In this review, we discuss the mechanisms of interaction of the GM and inflammation in AP and factors that can deteriorate or even cure both, including some traditional Chinese medicine treatments, to provide new methods for studying AP pathogenesis and developing therapies.
Article
Full-text available
Acute pancreatitis (AP) continues to present a substantial burden to patients and healthcare personnel. Despite its occasionally severe progression and high mortality rate, there is no specific therapy that could be routinely applied in patients with AP. Here, we review treatment possibilities in AP, describe how the treatment approaches have changed in pancreatic cancer as an analogy, and point out potential causes for the failure of clinical trials on AP. We highlight that instead of attempting to discover generalized treatment options that could be used in any AP patient, it is time for a paradigm shift in the treatment of AP, which would help to focus more on individual patients or specific patient subpopulations when designing clinical trials and therapeutic approaches (similarly as in pancreatic cancer). Since the recruitment of specific patient subpopulations with AP could take excessive time if clinical centers work separately, the development of precision medicine in AP would require to establish an expert committee, e.g., Pancreatitis Precision Medicine Interest Group, which could organize and coordinate the activities of the joined centers. With the joined forces of expert clinicians and leading centers, a new era could start in the treatment of AP, in which personalized treatment options could be discovered and introduced to efficiently reduce the burden of the disease on patients and healthcare workers.
Article
Full-text available
Acute pancreatitis (AP) is a disease characterized by local and systemic inflammation with an increasing incidence worldwide. Receptor-interacting serine/threonine protein kinase 3 (RIPK3), mixed-lineage kinase domain-like protein (MLKL), and innate immune cell macrophages have been reported to be involved in the pathogenesis of AP. However, the mechanisms by which RIPK3 and MLKL regulate pancreatic injury, as well as the interactions between injured pancreatic acinar cells and infiltrating macrophages in AP, remain poorly defined. In the present study, experimental pancreatitis was induced in C57BL/6J, Ripk3-/- and Mlkl-/- mice by cerulein plus lipopolysaccharide in vivo, and primary pancreatic acinar cells were also isolated to uncover cellular mechanisms during cerulein stimulation in vitro. The results showed that MLKL and its phosphorylated protein p-MLKL were upregulated in the pancreas of the mouse AP model and cerulein-treated pancreatic acinar cells, independent of its canonical upstream molecule Ripk3, and appeared to function in a cell death-independent manner. Knockout of Mlkl attenuated AP in mice by reducing the polarization of pancreatic macrophages toward the M1 phenotype, and this protective effect was partly achieved by reducing the secretion of CXCL10 from pancreatic acinar cells, whereas knockout of Ripk3 did not. In vitro neutralization of CXCL10 impaired the pro-M1 ability of the conditioned medium of cerulein-treated pancreatic acinar cells, whereas in vivo neutralization of CXCL10 reduced the polarization of pancreatic macrophages toward M1 and the severity of AP in mice. These findings suggested that targeting the MLKL-CXCL10-macrophage axis might be a promising strategy for the treatment of AP.
Article
Full-text available
Glutathione peroxidase 4 (GPX4)-dependent ferroptosis in pancreatic acinar cells plays a critical role in acute pancreatitis (AP). However, potential upstream regulators of GPX4 are not well defined. Here, we observed a marked reduction in acinar GPX4 expression and ferroptotic cell death in mice with cerulein-induced AP. To determine the critical factors involved in acinar cell ferroptosis, pancreas transcriptome data from an AP mouse model were analyzed and overlapped with predicted transcription factors of Gpx4, and an upregulated transcription factor active protein 1 (AP-1) protein, Jun, was identified. The administration of a specific ferroptosis inhibitor liproxstatin-1 alleviated AP pathology and significantly decreased Jun levels. Bioinformatic analysis indicated that the Gpx4 promoter contains a putative AP-1 binding site. Jun binds directly to the Gpx4 promoter and inhibits Gpx4 transcription under pancreatic conditions. AP-1 inhibition by a selective inhibitor SR11302 reversed GPX4 reduction and ameliorated AP pathology in a GPX4-dependent manner. Collectively, our study demonstrates that the downregulation of GPX4 by AP-1 is critical in the aggravation of acinar cell ferroptosis during the progression of AP. Strategies targeting the AP-1/GPX4 axis may be potentially effective for the prevention and treatment of AP.
Article
Importance: The incidence of hypertriglyceridemia-associated acute pancreatitis (HTG-AP) is increasing. Plasmapheresis is theoretically effective in removing triglyceride from plasma, but whether it confers clinical benefits is unclear. Objective: To assess the association between plasmapheresis and the incidence and duration of organ failure among patients with HTG-AP. Design, setting, and participants: This is an a priori analysis of data from a multicenter, prospective cohort study with patients enrolled from 28 sites across China. Patients with HTG-AP were admitted within 72 hours from the disease onset. The first patient was enrolled on November 7th, 2020, and the last on November 30th, 2021. The follow-up of the 300th patient was completed on January 30th, 2022. Data were analyzed from April to May 2022. Exposures: Receiving plasmapheresis. The choice of triglyceride-lowering therapies was at the discretion of the treating physicians. Main outcomes and measures: The primary outcome was organ failure-free days to 14 days of enrollment. Secondary outcomes included other measures for organ failure, intensive care unit (ICU) admission, duration of ICU and hospital stays, incidence of infected pancreatic necrosis, and 60-day mortality. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) analyses were used to control potential confounders. Results: Overall, 267 patients with HTG-AP were enrolled (185 [69.3%] were male; median [IQR] age, 37 [31-43] years), among whom 211 underwent conventional medical treatment and 56 underwent plasmapheresis. PSM created 47 pairs of patients with balanced baseline characteristics. In the matched cohort, no difference was detected concerning organ failure-free days between patients undergoing plasmapheresis or not (median [IQR], 12.0 [8.0-14.0] vs 13.0 [8.0-14.0]; P = .94). Moreover, more patients in the plasmapheresis group required ICU admission (44 [93.6%] vs 24 [51.1%]; P < .001). The IPTW results conformed to the results from the PSM analysis. Conclusions and relevance: In this large multicenter cohort study of patients with HTG-AP, plasmapheresis was commonly used to lower plasma triglyceride. However, after adjusting for confounders, plasmapheresis was not associated with the incidence and duration of organ failure, but with increased ICU requirements.
Article
Background: While transient bacteremia is common during dental and endoscopic procedures, infections developing during sterile diseases like acute pancreatitis (AP) can have grave consequences. Here we examined how impaired bacterial clearance may cause this transition. Methods: Blood samples of AP patients, normal controls, and rodents with pancreatitis or those administered different non-esterified fatty acids (NEFA) were analyzed for albumin unbound NEFA, microbiome, and inflammatory cell injury. Macrophage uptake of unbound-NEFA using a novel coumarin tracer were done, and the downstream effects, NEFA-membrane phospholipid (phosphatidylcholine; PC) interactions were studied on isothermal titration calorimetry (ITC) RESULTS: Infected AP patients had higher circulating unsaturated NEFA, unbound-NEFA including linoleic acid (LA) and oleic acid (OA), higher bacterial 16S DNA, mitochondrial DNA, altered beta-diversity, enrichment in Pseudomonadales and increased annexin V positive myeloid (CD14) and CD3 positive T cells on admission. These, and increased circulating dead inflammatory cells were also noted in rodents with unbound unsaturated-NEFA. ITC showed progressively stronger unbound-LA interactions with aqueous media, PC, cardiolipin and albumin. Unbound-NEFA were taken into protein free membranes, cells, mitochondria, inducing voltage dependent anion channel oligomerization, reducing ATP, and impairing phagocytosis. These were reversed by albumin. In-vivo unbound-LA, OA increased bacterial loads and impaired phagocytosis, causing infection. LA, OA were more potent for these amphipathic interactions than the hydrophobic palmitic acid. Conclusions: Release of stored LA, OA can increase their circulating unbound levels and cause amphipathic liponecrosis of immune cells via uptake by membrane phospholipids. This impairs bacterial clearance and causes infection during sterile inflammation.
Article
Objective: There is an urgent need for safe and targeted interventions to mitigate post-ERCP pancreatitis (PEP). Calcineurin inhibitors (CnIs) offer therapeutic promise as calcineurin signaling within acinar cells is a key initiating event in PEP. In previous proof-of-concept studies using experimental models, we showed that concurrent intra-pancreatic ductal administration of the CnIs, tacrolimus (Tac) or cyclosporine A (CsA) with the ERCP radiocontrast agent (RC) prevented PEP. To translate this finding clinically, we investigated potential toxic effects of intraductal delivery of a single-dose RC-CnI formulation on endocrine pancreas function and systemic toxicities in a preclinical PEP model. Methods: C57BL/6J mice underwent ductal cannulation and received a single, intra-pancreatic ductal infusion of RC or RC with Tac or CsA (treatment groups) or underwent ductal cannulation without infusion ('sham' group). To assess endocrine function, intraperitoneal glucose tolerance test (IPGTT) was performed at two days before infusion and on day 2 and 14 post-surgery. To evaluate off-target tissue toxicities, renal and hepatic function-related parameters including blood urea nitrogen, plasma creatinine, potassium, aspartate aminotransferase, alanine aminotransferase, and total bilirubin were measured at the same time-points as IPGTT. Histological and biochemical indicators of pancreas injury and inflammation were also evaluated. Results: No abnormalities in glucose metabolism, hepatic or renal function were observed on day 2 or 14 in mice administered with intraductal RC or RC with Tac or CsA. Conclusion: Intraductal delivery of RC-CnI formulation was safe and well-tolerated with no significant acute or subacute endocrine or systemic toxicities, underscoring its clinical utility to prevent PEP.
Article
Hypertriglyceridaemia (HTG) is a metabolic disorder, defined when serum or plasma triglyceride concentration (seTG) is >1.7 mM. HTG can be categorized as mild to very severe groups based on the seTG value. The risk of acute pancreatitis (AP), a serious disease with high mortality and without specific therapy, increases with the degree of HTG. Furthermore, even mild or moderate HTG aggravates AP initiated by other important aetiological factors, including alcohol or bile stone. This review briefly summarizes the pathophysiology of HTG, the epidemiology of HTG‐induced AP and the clinically observed effects of HTG on outcomes of AP. Our main focus is to discuss the pathophysiological mechanisms linking HTG to AP. HTG is accompanied by an increased serum fatty acid (FA) concentration, and experimental results have demonstrated that these FAs have the most prominent role in causing the consequences of HTG during AP. FAs inhibit mitochondrial complexes in pancreatic acinar cells, induce pathological elevation of intracellular Ca2+ concentration, cytokine release and tissue injury, and reduce the function of pancreatic ducts. Furthermore, high FA concentrations can induce respiratory, kidney, and cardiovascular failure in AP. All these effects may contribute to the observed increased AP severity and frequent organ failure in patients. Importantly, experimental results suggest that the reduction of FA production by lipase inhibitors can open up new therapeutic options of AP. Overall, investigating the pathophysiology of HTG‐induced AP or AP in the presence of HTG and determining possible treatments are needed.
Article
In this account of the 2022 Palade Medal Lecture, an attempt is made to explain, as simply as possible, the most essential features of normal physiological control of pancreatic enzyme secretion, as they have emerged from more than 50 years of experimental work. On that basis, further studies on the mechanism by which acute pancreatitis is initiated are then described. Calcium ion signaling is crucially important for both the normal physiology of secretion control as well as for the development of acute pancreatitis. Although acinar cell processes have, rightly, been central to our understanding of pancreatic physiology and pathophysiology, attention is here drawn to the additional critical influence of calcium signaling events in stellate and immune cells in the acinar environment. These signals contribute significantly to the crucially important inflammatory response in acute pancreatitis.
Article
Acute pancreatitis (AP) is a disease characterized by an acute inflammatory phase followed by a convalescent phase. Diabetes mellitus (DM) was historically felt to be a transient phenomenon related to acute inflammation; however, it is increasingly recognized as an important late and chronic complication. There are several challenges that have prevented precisely determining the incidence rate of DM after AP and understanding the underlying mechanisms. The DREAM (Diabetes RElated to Acute Pancreatitis and its Mechanisms) Study is a prospective cohort study designed to address these and other knowledge gaps to provide the evidence needed to screen for, prevent, and treat DM after AP. In the following article, we summarize literature regarding the epidemiology of DM after AP and provide the rationale and an overview of the DREAM study.