ArticlePDF AvailableLiterature Review

Abstract and Figures

Since the discovery of the first trypsinogen mutation in families with hereditary pancreatitis, pancreatic genetics has made rapid progress. The identification of mutations in genes involved in the digestive protease–antiprotease pathway has lent additional support to the notion that pancreatitis is a disease of autodigestion. Clinical and experimental observations have provided compelling evidence that premature intrapancreatic activation of digestive proteases is critical in pancreatitis onset. However, disease course and severity are mostly governed by inflammatory cells that drive local and systemic immune responses. In this article, we review the genetics, cell biology, and immunology of pancreatitis with a focus on protease activation pathways and other early events.
Content may be subject to copyright.
Genetics, Cell Biology, and Pathophysiology of Pancreatitis
1
Medical Department II, University Hospital, LMU, Munich, Germany;
2
Department of Medicine A, University Medicine
Greifswald, Greifswald, Germany;
3
Institute for Translational Medicine, University of Pécs, Pécs, Hungary; and
4
Center for
Exocrine Disorders, Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental
Medicine, Boston, Massachusetts
Since the discovery of the rst trypsinogen mutation in
families with hereditary pancreatitis, pancreatic genetics
has made rapid progress. The identication of mutations
in genes involved in the digestive proteaseantiprotease
pathway has lent additional support to the notion that
pancreatitis is a disease of autodigestion. Clinical and
experimental observations have provided compelling evi-
dence that premature intrapancreatic activation of diges-
tive proteases is critical in pancreatitis onset. However,
disease course and severity are mostly governed by in-
ammatory cells that drive local and systemic immune
responses. In this article, we review the genetics, cell
biology, and immunology of pancreatitis with a focus on
protease activation pathways and other early events.
Keywords: Trypsinogen; Pancreatitis; Genetics; Inammation;
Cell Death.
Pancreatitis is the leading cause for gastrointestinal
disease-related hospital admissions and it is associ-
ated with considerable morbidity, mortality, and socioeco-
nomic burden.
1
Recent years have shed light on the
pathophysiology of pancreatitis, opening up new avenues
for causal treatment. In this review article, we dissect the
complexity of premature protease activation and its effect
on local and systemic inammation in pancreatitis.
Genetics of Pancreatitis
Acute pancreatitis (AP), recurrent AP (RAP), and chronic
pancreatitis (CP) form a disease continuum.
2
The progres-
sion of a sentinel attack of AP to RAP and eventually to CP is
often driven by chronic alcohol consumption or genetic risk
factors. Genetic risk for RAP and CP overlaps, whereas ge-
netic studies in AP are difcult to interpret in the absence of
adequate follow-up that can exclude RAP and CP cases.
Most pancreatitis risk genes code for digestive proteases,
a trypsin inhibitor, or other proteins highly expressed in the
pancreas. Functional studies have classied the various
mutations and other genetic alterations into pathologic
pathways driving pancreatitis onset and progression. We
discuss the trypsin-dependent, mis-foldingdependent, and
ductal pathways of pancreatitis risk.
Trypsin-Dependent Pathway of Genetic
Risk in CP
Pancreatic acinar cells secrete digestive proteases in
inactive precursor forms that are ushed from the ductal
system in a sodium bicarbonaterich uid. Trypsinogen, the
precursor to trypsin, becomes activated by the serine pro-
tease enteropeptidase in the duodenum.
3
Trypsin activates
chymotrypsinogens, proelastases, and procarboxypeptidase
B1 (CPB1), whereas activation of procarboxypeptidases A1
(CPA1) and A2 (CPA2) requires the concerted action of
trypsin and chymotrypsin C (CTRC).
4
Trypsinogen also can
be activated by trypsin, and this process is called autoacti-
vation.
3
Premature, intrapancreatic activation of trypsin-
ogen can occur by autoactivation or can be catalyzed by the
lysosomal cysteine protease cathepsin B. Protective mech-
anisms that prevent trypsinogen activation in the pancreas
include trypsin inhibition by the serine protease inhibitor
Julia Mayerle
1,2
Matthias Sendler
2
Eszter Hegyi
3
Georg Beyer
1
Markus M. Lerch
2
Miklós Sahin-Tóth
4
Abbreviations used in this paper: AP, acute pancreatitis; AP1, activator
protein 1; CASR, calcium-sensing receptor; CCK, cholecystokinin; CEL,
carboxyl ester lipase; CEL-HYB, hybrid carboxyl ester lipase allele; CFTR,
cystic brosis transmembrane conductance regulator; CLDN2, claudin 2;
CP, chronic pancreatitis; CPA1, procarboxypeptidase A1; CPA2, pro-
carboxypeptidase A2; CPB1, procarboxypeptidase B1; CTRC, chymo-
trypsin C; DAMP, damage-associated molecular pattern; ER, endoplasmic
reticulum; EV, endocytic vacuole; GWAS, genomewide association study;
HP, hereditary pancreatitis; IKK, inhibitor of nuclear factor kB kinase; IL,
interleukin; MODY8, maturity-onset diabetes of the young type 8; MyD88,
myeloid differentiation primary response 88; NET, neutrophil extracellular
trap; NFkB, nuclear factor klight-chain enhancer of activated B cells;
NLRP3, nucleotide-binding oligomerization domain-like receptor protein
3; OR, odds ratio; PRSS1, cationic trypsinogen; PRSS2, anionic trypsin-
ogen; RAP, recurrent acute pancreatitis; RIP, receptor-interacting protein;
ROS, reactive oxygen species; SPINK1, serine protease inhibitor Kazal
type 1; TLR, Toll-like receptor; TNF, tumor necrosis factor.
Most current article
© 2019 by the AGA Institute
0016-5085/$36.00
https://doi.org/10.1053/j.gastro.2018.11.081
Gastroenterology 2019;156:19511968
PANCREATITIS
Kazal type 1 (SPINK1) and trypsinogen degradation by
CTRC and cathepsin L.
57
Although the principal action of
CTRC is to promote trypsinogen degradation, it also enhances
trypsinogenactivationbyprocessing the trypsinogen activa-
tion peptide to a shorter form, which is more sensitive to
trypsin-mediated activation
79
(Figure 1). As discussed below,
certain trypsinogen mutations can hijack this mechanism and
thereby stimulate trypsinogen activation to a pathologic extent.
Human genetic studies strongly support trypsinogen autoac-
tivation and CTRC-dependent trypsinogen degradation as key
mechanisms determining intrapancreatic trypsin activity,
whereas similarly compelling genetic evidence for the role of
cathepsins B and L has been lacking.
10
Cationic Trypsinogen Mutations
Mutations in human cationic trypsinogen (PRSS1)cause
autosomal dominant hereditary pancreatitis (HP) with
incomplete penetrance or act as risk factors in sporadic
CP.
7,11
Approximately 90% of PRSS1-mutationpositive HP
families carry the p.N29I, p.R122C, or p.R122H mutation in
the heterozygous state. Mechanistically, the p.R122C and
p.R122H mutations prevent CTRC-mediated trypsinogen
degradation.
9
The p.N29I mutation has multiple distinct ef-
fects on trypsinogen biochemistry, the combination of which
markedly increases trypsinogen autoactivation. These effects
include an increase in N-terminal processing, decreased
CTRC-dependent degradation, and a slightly increased pro-
pensity for autoactivation.
9
The p.A16V variant sensitizes the
activation peptide of trypsinogen to CTRC-mediated pro-
cessing, which in turn enhances autoactivation.
8,9
Pathologic
trypsin levels generated by mutation p.A16V are lower than
those seen with the p.R122H variant, which explains the
decreased penetrance of the p.A16V variant. More recently,
mutation p.P17T was found to exhibit characteristics that
were similar to those of p.A16V.
12
Rare mutations affecting
the activation peptide of cationic trypsinogen (p.D19A,
p.D21A, p.D22G, p.K23R, and p.K23_I24insIDK) robustly
stimulate autoactivation independently of CTRC.
1315
Cell
culture experiments have indicated that these activation
peptide mutants are secreted poorly because of intracellular
activation and degradation, which can lead to cellular stress
and consequent acinar cell death.
16
Taken together, PRSS1
mutations stimulate activation of cationic trypsinogen by
decreasing CTRC-dependent trypsinogen degradation,
increasing CTRC-mediated processing of the activation pep-
tide, or directly stimulating autoactivation. Genomewide as-
sociation studies (GWASs) have identied a commonly
occurring haplotype in the PRSS1 and anionic trypsinogen
(PRSS2) locus that slightly decreases CP risk (odds ratio [OR]
1.5), with a more pronounced effect in alcoholic CP.
1719
A
variant (c.204C>A) that lies in the promoter region of
PRSS1 and decreases trypsinogen expression appears to be
responsible for this small protective effect.
20
SPINK1 Mutations
The association between the most common p.N34S
SPINK1 variant and CP was rst described by a candidate
gene study in 2000.
21
A meta-analysis reported a carrier
frequency of 9.7% in patients with CP and 1% in controls
with an average OR of 11, making the p.N34S the clinically
most signicant risk factor for CP.
22
When considering
European populations only, p.N34S increased CP risk by
approximately 10-fold.
23
Although several studies have
attempted to identify the functional effect of p.N34S and its
associated haplotype, the molecular mechanism underlying
CP risk remains unclear. Neither p.N34S nor any of the 4
linked intronic variants affect trypsin inhibitory function or
cellular expression of SPINK1.
2427
Interestingly, in pancre-
atic cancer cell lines carrying the heterozygous p.N34S
variant, decreased expression of the mutant allele was
observed compared with the wild-type allele.
28
The in-
vestigators suggested that the c.4141G>T variant or a
hitherto unknown variant located in the 50region of the gene
might be responsible for the decreased expression of the
p.N34S allele. The second most frequently reported SPINK1
haplotype in CP contains the c.215G>A promoter variant
and the c.194þ2T>C variant in intron 3.
21,29
This haplotype
was observed more frequently in East Asia than in Europe.
7
Functional studies have shown that the c.194þ2T>C
variant causes skipping of exon 3, which results in dimin-
ished SPINK1 expression.
27,30,31
However, the c.215G>A
variant increases promoter activity, which might mitigate the
effect of the c.194þ2T>C mutation and allow for some re-
sidual SPINK1 expression even in homozygous carriers.
32,33
A
large number of rare or private alterations in SPINK1 have
been found in CP, which cause loss of SPINK1 function by
various mechanisms.
7
Protective PRSS2 Variant
Although PRSS1 and PRSS2 share 90% identity at the
amino acid level and PRSS2 rapidly auto-activates, no path-
ogenic PRSS2 variants have been identied in HP or sporadic
CP.
34,35
The absence of PRSS2 mutations in CP could be due
to the more effective CTRC-mediated degradation of anionic
trypsinogen, which would prevent intrapancreatic activation
of the enzyme even if it were mutated.
36
However, a pro-
tective variant p.G191R with a w3- to 6-fold effect and
approximately 5% population frequency was discovered.
35,37
The mutation introduces a new trypsin cleavage site into
anionic trypsinogen, which increases autocatalytic proteolysis
and inactivation.
35
CTRC Mutations
Direct DNA sequencing of the CTRC gene in patients with
nonalcoholic CP showed heterozygous mutations in 4% of
patients that increased CP risk by 5-fold on average.
38,39
The
mutations cause loss of CTRC function by various mecha-
nisms, which include defective secretion from mis-folding,
resistance to trypsin-mediated activation, catalytic de-
ciency, or increased degradation by trypsin.
40,41
Considering
the clinically signicant variants, p.A73T exhibits a severe
secretion defect, p.K247_R254del is inactive and prone to
degradation, p.R254W is degraded by trypsin, and p.V235I
has partly decreased activity.
40
Subsequent studies reported
a frequent p.G60¼variant found in approximately 30% of
patients with CP.
4245
The heterozygous p.G60¼increases
1952 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
the risk of CP by 2.5-fold, whereas the homozygous state
increases the risk by 10-fold.
43,45
The variant is associated
with decreased CTRC mRNA expression (GTEx Portal),
possibly because of altered pre-mRNA splicing.
CTRB1CTRB2 Locus Inversion
A recent European GWAS identied a large inversion at
the CTRB1CTRB2 locus that modestly (OR 1.35) modies the
risk for alcoholic and nonalcoholic CP.
19
The inversion
changes the expression ratio of the CTRB1 and CTRB2
chymotrypsin isoforms in such a manner that protective
trypsinogen degradation is increased and CP risk is decreased.
In China, the reported population frequency of the inverted
(major) allele is 99.6%; thus, the allele is virtually xed and
does not contribute to CP risk.
46
A mouse model with genetic
deletion of the major mouse chymotrypsin CTRB1 exhibited
increased intra-acinar trypsin activation and more severe
pancreatitis induced by the secretagogue cerulein.
47
These
observations provided the rst in vivo proof for the protective
role of chymotrypsin-mediated trypsinogen degradation
against pancreatitis.
Mis-foldingDependent Pathway of
Genetic Risk in CP
More recently, an alternative pathomechanism seem-
ingly unrelated to premature intrapancreatic trypsinogen
activation has been identied, in which mutation-induced
mis-folding and consequent endoplasmic reticulum (ER)
stress lead to acinar cell damage and pancreatitis.
48
Mis-foldingAssociated PRSS1 Mutations
In 2009, a subset of PRSS1 variants was found to cause
decreased secretion, intracellular retention, and increased
ER stress markers, as judged by in vitro cell culture ex-
periments.
49
These PRSS1 mutations occur rarely and are
mostly associated with sporadic disease (eg, p.C139F,
p.C139S, p.G208A), but also have been found in HP families
with incomplete penetrance (p.L104P, p.R116C).
48
Variant
p.G208A is prevalent in East Asia (4% of CP cases) and was
detected in Europe only in a single case thus far.
50,51
Mis-foldingAssociated CPA1 Mutations
A candidate gene study in 2013 reported that mutations
in the CPA1 gene are associated with CP (OR w25), espe-
cially with early-onset disease (OR w80).
52
The vast ma-
jority of pathogenic CPA1 variants occur with low frequency
and are mostly found in sporadic CP. The p.S282P variant
was described in 2 HP families.
53
Pathogenic CPA1 variants
cause proenzyme mis-folding, resulting in a secretion defect,
intracellular retention, and ER stress.
52,53
In contrast to
CPA1, variants of CPB1 and CPA2 are not associated with
CP.
54
Interestingly, ER stress-inducing CPA1 and CPB1 var-
iants were over-represented in patients with pancreatic
cancer without a clinical history of RAP or CP.
55
Most of
these variants caused premature truncation and did not
overlap with those found in CP. A mouse model for the mis-
foldingdependent pathway was described recently. This
study showed that CPA1 N256K knock-in mice harboring the
most frequent p.N256K human CPA1 mutation develop
spontaneous and progressive CP and exhibit signs of ER
stress in their pancreas.
56
Mis-foldingAssociated CEL Mutations and
CEL-HYB Allele
Single-nucleotide deletions in the last exon of the CEL
gene encoding carboxyl ester lipase cause maturity-onset
diabetes of the young type 8 (MODY8).
57
The deletions
alter the reading frame of the C-terminal variable number
tandem repeat sequence, resulting in CEL proteins with
unnatural extensions that are prone to aggregation.
58,59
The
exocrine dysfunction in MODY8 is in all likelihood caused by
mis-foldinginduced ER stress and consequent acinar cell
loss. A hybrid CEL allele (CEL-HYB1) formed between CEL
Figure 1. Genetic risk factors associated with the trypsin-dependent pathologic pathway. See text for details.
May 2019 Genetics and Pathophysiology of Pancreatitis 1953
PANCREATITIS
and its neighboring pseudogene CELP was over-represented
in idiopathic CP by approximately 5-fold vs the average
population frequency of 0.5%1%.
60
In cell culture experi-
ments, the hybrid protein was secreted poorly because of
intracellular retention, suggesting that the CEL-HYB1 variant
can increase CP risk by the mis-foldingdependent pathway.
A second hybrid CEL allele (CEL-HYB2) that does not asso-
ciate with CP was described in Asian populations.
61
Inter-
estingly, the CEL protein carries blood group antigens and a
GWAS in 2015 indicated that fucosyl-transferase 2 nonse-
cretor status and blood group B are risk factors for CP.
62
Although other studies in ethnically mixed cohorts failed
to replicate this association,
63,64
with the exception of
azathioprine-induced pancreatitis in patients with inam-
matory bowel disease,
65
it is still interesting to speculate
that the observed effects might have been due to changes in
CEL folding or trafcking.
Ductal Pathway of Genetic Risk in CP
CFTR Variants
The cystic brosis transmembrane conductance regu-
lator (CFTR) is a cyclic adenosine monophosphate
regulated chloridebicarbonate channel localized to the
apical plasma membrane of epithelial cells
66
(Figure 2).
CFTR mutations disrupt channel activity or affect membrane
levels and are associated with various phenotypes, ranging
from asymptomatic state to multiorgan symptoms leading
to the diagnosis of cystic brosis in homozygous carriers of
severe mutations. Observations that heterozygous and
compound heterozygous CFTR mutations are associated
with CP were reported by 2 studies in 1998.
67,68
In the rst
analysis of the entire CFTR coding region, the frequency of
abnormal CFTR alleles in patients with CP was 18.6%
compared with 9.2% in controls.
69
More recent large cohort
analyses have corroborated the pathogenic role of CFTR
variants in CP, although the effect and frequency of CFTR
variants was less pronounced than reported previ-
ously.
39,70,71
Heterozygous carrier status of the severe
p.F508del mutation confers a small risk for CP (OR 2.5),
whereas the mild p.R117H mutation increases risk by
approximately 4-fold. Compound heterozygous state for 1
severe and 1 mild CFTR allele represents strong risk for CP
and can be considered causative.
70
The role of common
polymorphic CFTR alleles (eg, T5, TG12) and the noncystic
brosiscausing, so-called bicarbonate-defective CFTR vari-
ants in CP remains controversial because the preponderance
of data does not support their association with CP.
66
Unlike
CFTR, variants in the solute-linked carrier 26 member 6 anion
transporter (SLC26A6) do not alter the genetic risk in CP.
72
CLDN2 Variants
GWASs of CP identied several single-nucleotide pep-
tides in the claudin 2 (CLDN2) and MORC4 locus to be
Figure 2. Genetic risk fac-
tors associated with the
ductal pathologic pathway.
See text for details.
1954 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
associated with CP risk.
17,19
The OR was approximately 2
and the effect was more pronounced in alcoholic CP. Within
this locus, CLDN2 seems to be the clinically relevant risk gene,
because it is expressed in pancreatic ducts at low levels as a
tight junction protein. It was proposed that CLDN2MORC4
variants might cause CLDN2 mis-localization. Additional
work is required to clarify the mechanism of action of this
risk locus and to conrm whether assignment to the ductal
pathway is appropriate (Figure 2).
CASR Variants
The calcium-sensing receptor (CASR) regulates calcium
homeostasis through parathyroid hormone secretion and
renal tubular calcium reabsorption. Functional CASR also is
expressed in the pancreas, including ductal cells where
CASR can respond to high calcium concentrations in the
juice by increasing ductal uid secretion, thereby preventing
stone formation and pancreatitis
73
(Figure 2). A US
population-based study failed to demonstrate the previously
anticipated association between CASR variants and the
SPINK1 p.N34S haplotype, but reported the p.R990G variant
increased CP risk, especially in subjects with moderate or
heavy alcohol consumption.
74
More recently, a French study
found over-representation of rare CASR coding variants in
idiopathic CP and a signicant association of the p.A986S
variant, but only in the homozygous state, with CP.
75
How-
ever, the previously reported association with the p.R990G
variant was not observed in this cohort. Taken together,
current evidence does not support a clear role for CASR
variants in CP pathogenesis.
In summary, human genetic data indicate that premature
activation or mis-folding of pancreatic proteases play a
central role in the onset of pancreatitis and progression to
CP (Supplementary Table 1).
Role of Proteases in Pathophysiology
and Cell Biology of Pancreatitis
Although genetic evidence for the involvement of the
proteaseantiprotease balance in the pathogenesis of
pancreatitis dates back only 2 decades and focuses mainly
on CP, pathophysiologic and biochemical investigations have
implicated this system for more a century. Because of the
lack of adequate animal models and the inability to keep
isolated pancreatic acinar cells in culture for long periods,
experimental studies have focused primarily on AP. The
relative importance of the pathways discussed below might
change with respect to etiology. It is our general under-
standing that these mechanisms also are relevant to CP,
although experimental evidence is mostly lacking.
Autodigestion by Pancreatic Proteases
The pathophysiologic concept of autodigestion was rst
developed by the Austrian pathologist Hans Chiari in Prague
more than 120 years ago. He claimed that pancreatitis was
caused and driven by the glandsdigestive properties.
76
Since then, the pathomechanism of premature activation
of pancreatic enzymes and its contribution to disease
severity and progression have captured the attention of
many pancreatologists. Bialek et al
77
rst reported that
protease activation during pancreatitis begins in the
exocrine pancreas and Hofbauer et al
78
reported that it
begins in a membrane-conned vesicular compartment and
parallels acinar cell damage. Although the fact that activa-
tion of digestive proteases is an early event during AP is
widely accepted, the question of where and through what
mechanism this process is initiated and whether it plays a
role in chronicity remain under debate.
Protease Activation During PancreatitisClues
From Mechanistic Studies
Role of Calcium for Intracellular Protease Activa-
tion. States of hypercalcemia, such as primary hyperpara-
thyroidism, are a risk factor for the development of AP in
humans and rats.
7981
Intracellular calcium concentrations
and compartmental distribution in acinar cells are tightly
regulated because calcium serves as a second messenger for
the physiologic release of digestive enzymes in response to
vagal nerve stimulation or humoral activation.
82,83
After
intraperitoneal treatment of rats with supramaximal doses
of cerulein, an analogue of cholecystokinin,
84
a time-
dependent disruption of the physiologic oscillating intra-
cellular calcium signal was observed in rat acini using the
Ca
2þ
-sensitive dye fura-2.
85
When using for the rst time
uorescent trypsin substrates that allow the subcellular
imaging, localization, and quantication of protease activa-
tion,
86
Krüger et al
87
found that a specic extended plateau
release of calcium at the apical pole of acinar cells is
necessary for premature trypsin activation to occur, which
is different from the calcium oscillations required for
enzyme secretion. These ndings were conrmed by others
who also found that acetylcholine- or cholecystokinin (CCK)-
induced intracellular protease activation was associated
with the formation of cytoplasmic vacuoles in acinar cells
that resemble those overserved in experimental pancreatitis
in vivo
88
and that inhibition of calcium release also inhibits
the formation of these vesicles.
89
Acinar cells can replace
calcium for magnesium in its stores and, when this is done,
not only the premature activation of proteases in vitro and
in vivo but also the severity of pancreatitis is signicantly
decreased.
90
Two ongoing clinical trials (1 in AP and 1 in
CP) are based on this observation. Orabi et al
91
took the
concept of calcium-dependent protease activation further by
showing that AP induced by supramaximal doses of the
muscarinic agonist carbachol can be abrogated by inhibition
of the intracellular calcium channel ryanodine receptor,
which is located at the basolateral membrane of acinar cells.
Therefore, the importance of spatial distribution of calcium
to the apical pole might be a specic feature of the cerulein
model. Interestingly, carbachol-induced protease activation
is more severe if cells are pretreated with ethanol. Calcium
signaling also could play a role in other forms of experi-
mental pancreatitis, such as bile acid-induced pancreatitis,
92
pressure-induced pancreatitis, and pancreatitis after endo-
scopic retrograde cholangiopancreatography.
93
Experi-
mental pancreatitis can be ameliorated by modulating
May 2019 Genetics and Pathophysiology of Pancreatitis 1955
PANCREATITIS
calcium release from intracellular stores or inux through
the plasma membrane by pharmacologic inhibition of
inositol triphosphate receptor (predominantly types 2 and
3) signaling
94,95
or calcium release-activated calcium
modulator 1.
96,97
The calcium-dependent protease activa-
tion also heavily depends on calcineurin, a calcium-activated
phosphatase, and its downstream signaling through the
transcription factor nuclear factor of activated T cells. In-
hibition of calcineurin by inhibitors or in mice lacking cal-
cineurin subunits causes decreased intracellular protease
activity in secretagogue or bile acidinduced pancreatitis,
without affecting vesicular transport.
98,99
Mechanisms of Protease Activation. Three major
concepts have been investigated over the past decades:
autoactivation, spatial redistribution, and fusion of zymogen
granules with other organelles and failure of protective
mechanisms.
Autoactivation of Trypsin. There is strong evidence
from human genetic studies indicating that autoactivation of
trypsinogen
100
causes CP in affected humans.
9
In contrast,
trypsinogen autoactivation is unimportant in experimental
cerulein-induced pancreatitis. Inhibition of active trypsin by
the reversible chemical inhibitor S124 could prevent trypsin
activity in experimental cerulein-induced AP, but had no ef-
fect on the generation of the trypsin activation peptide (TAP)
or active trypsin after washout of S124, thus indicating that
hydrolysis of trypsinogen to trypsin appeared in a trypsin-
independent fashion.
101
Activation by Lysosomal Proteases. Inhibition of the
lysosomal hydrolase cathepsin B by the cysteine protease
inhibitor E-64d leads to a signicant decrease in trypsin
activity and TAP formation, indicating that the trypsin
activation in response to cerulein depends on cathepsin
B.
101103
Similarly, in cathepsin B knockout mice, trypsin
activation is signicantly inhibited after cerulein adminis-
tration.
104
In the past, it was thought that cathepsin B
105
and trypsinogen under physiologic conditions are not
located in the same cell organelles (zymogen granules for
exocytosis vs lysosomes for degradation of content of
endosomes and autophagosomes). In 1998, investigators
showed in subcellular fractions of cerulein-treated acini a
colocalization of cathepsin B and digestive enzymes,
including trypsin and TAP, in heavy fractions
78
containing
zymogen granules and lysosomes early in the disease course
and later a shift of trypsin and cathepsin B activity to the
cytosol.
106
This conrmed earlier ndings from in vivo
studies that indicated a fusion of zymogen-containing vac-
uoles with lysosomes in secretagogue-, duct obstruction-, or
diet-induced AP.
107112
Intracellular activation of proteases
other than trypsin, such as chymotrypsin and carboxypep-
tidase B, also depend on non-physiologic colocalization with
other cell components, but are independent of cathepsin
B.
113
Mis-sorting in the exocrine machinery, secretion
blockage, and reuptake of previously secreted proteases by
endocytosis have been described under experimental con-
ditions. The fact that an acidic pH, as found in lysosomes,
enhances secretagogue-dependent zymogen activation sup-
ports the fusion hypothesis.
114
A very recent study reported
that CCK or ethanol treatment depletes acinar cells of
syntaxin 2, a key regulator of apical exocytosis, thus leading
to increased basolateral exocytosis and formation of auto-
lysosomes mediated by syntaxins 3 and 4, in which tryp-
sinogen activation takes place.
115
Inhibition of syntaxin-4
mediated basolateral exocytosis in experimental pancrea-
titis decreases disease severity.
116
Another concept in-
troduces endocytic vacuoles (EVs) as the site of intracellular
trypsinogen activation. These occur under physiologic and
pathophysiologic conditions after compound exocytosis of
zymogen granules.
117
EV formation is calcium depen-
dent.
118
Their content is acidic and calcium rich and, after
supramaximal CCK or taurocholate stimulation, trypsin ac-
tivity within these post-exocytic structures can be visualized
using uorescent dyes.
119
During pancreatitis, EVs are
larger than normal and tend to fuse with the plasma
membrane or even rupture, discharging active trypsin into
the cytosol or extracellular space. This instability is believed
to be caused by disruption of otherwise protective actin
laments surrounding the EV.
120
However, rupture of EVs is
independent of trypsin or cathepsin B activity. Secretory
blockage can contribute to these events. In experimental
pancreatitis, vesicle-associated membrane protein-8medi-
ated secretion is impaired because of a loss of early endo-
somal proteins, resulting in retention of trypsinogen and
transformation to active trypsin in a cathepsin-dependent
manner. Knockout of vesicle-associated membrane
protein-8 protects against pancreatitis and restoration of
early endosomal trafcking decreases severity of pancrea-
titis.
121,122
Vesicular trafcking is regulated in a calcium-
dependent manner.
123,124
Loss of Trypsin Inhibitors. Little is known about the role
of failing protective mechanisms for protease activation in the
early phase of pancreatitis. The most potent cellular trypsin
inhibitor is SPINK1. Although SPINK1 mutations are among
the most common genetic risk factors for the development of
recurrent AP and CP, thus far, none of the described
mutations seemed to impair SPINK function and therefore do
not explain the increased risk for pancreatitis.
2427
In mice
carryingaheterozygousSPINK3deletion,asignicant
decrease of functional SPINK does not lead to the develop-
ment of spontaneous pancreatitis or more severe disease
after supramaximal cerulein administration compared with
wild-type controls.
125
The fact that mice with a homozygous
SPINK3 deletion develop pancreatic atrophy and that this
phenotype can be rescued by transgenic expression of rat
pancreatic secretory trypsin inhibitor 1
125
point toward a
role of trypsin inhibitors during pancreatic development but
do not explain that role in pancreatitis.
In conclusion, the current cumulative evidence suggests
a cathepsin Bdependent mechanism of protease activation
in experimental pancreatitis.
Cell Death Cause or Consequence of Protease
Activation?
Premature intracellular protease activation in acinar
cells leads to cell injury. Type of cell death, be it necrosis,
apoptosis, autophagy, necroptosis, or pyroptosis, de-
termines disease severity.
126
Necrosis is understood as an
1956 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
unregulated response to damage. In animal models of AP,
approximately 1%5% of acinar cells undergo apoptosis
and severity of pancreatitis is inversely correlated to the
rate of apoptosis.
127
Macroautophagy is a multistep,
lysosomal-driven, adaptive process by which cells degrade
cytoplasmic organelles and long-lived protein.
128
Pancrea-
titis presents with impaired autophagic ux evident by
vacuole accumulation.
129
Currently, it is debated whether
impaired autophagy stimulates cell death through accumu-
lation of damaged mitochondria mediating an inammatory
response through a reactive oxygen species (ROS)-dependent
mechanism as in lysosome-associated membrane protein
2
130
or Atg7-knockout animals
131
or whether autophagy
prevents an inammatory response as in Atg5-knockout
mice.
132,133
The regulated process of necrosis is termed
necroptosis and is triggered by tumor necrosis factor (TNF),
TNF-related apoptosis-inducing ligand, Fas ligand, interferon
type 1, and Toll-like receptors (TLRs), which are released in
theearlyphaseofAPinresponsetoproteaseactivation.
134
Receptor-interacting protein 1 (RIP1) and RIP3 form a
phosphorylated complex, the necrosome, and phosphorylate
mixed-lineage kinase domain-like, resulting in membrane
rupture. Forty percent of cells undergo necroptosis in
pancreatitis and RIP3 deletion or treatment with necrostatin
ameliorates pancreatitis.
135,136
Necroptosis releases damage-
associated molecular patterns (DAMPs) and those will acti-
vate the nucleotide-binding oligomerization domain-like
receptor protein 3 (NLRP3) pathway, resulting in pyropto-
sis.
137
Pyroptosis is an innate immune sensing mechanism
with poorly understood upstream signaling. Nevertheless,
some interesting inhibitors are known, such as lactate,
b-hydroxybutyrate, and aspartate. The term pyroptosis de-
scribes activation of the inammasome through NLRP3. The
inammasome is a macroscopic cytosolic protein complex
that proteolytically cleaves interleukin (IL) 1band pro-IL18
and releases high mobility group box 1 protein. NLRP3 acti-
vation requires lysosomal rupture and cathepsin release,
calcium inux, and mitochondria-derived ROS production
and thus is closely linked to pancreatitis.
138,139
However,
NLRP3 expression is restricted to innate immune cells.
140
Cell death pathways in pancreatitis intersect. Caspase 3
activation can induce not only apoptosis but also pyroptosis.
Necroptosis can shift to pyroptosis by caspase 8 activation
and necroptosis activates pyroptosis.
141
Currently, we have
not understood why some of our patients deteriorate and
develop a necrotizing course of pancreatitis. A shift of
regulated cell death from apoptosis to pyroptosis and
stimulation of necroptosis might explain this observation.
Inhibition of pyroptosis, for example, by using Ringers
lactate for volume resuscitation
142
or inhibition of nec-
roptosis by necrostatin,
143
might well be a way forward in
the treatment of pancreatitis.
An interesting question is whether cell death is a result
of premature protease activation or a consequence of
inammation. The answer is ambiguous: Talukdar et al
106
reported a direct effect on lysosomal stability mediated by
active trypsin and lysosomal rupture leading to the release
of cathepsins into the cytosol, causing dose-dependently
apoptosis or necrosis. Our group showed that inhibition of
protease activation, especially trypsin by specic inhibitors,
results in a decreased rate of apoptosis but does not affect
necrosis.
144
Thus, cell death is a result of intracellular pro-
tease activation, but this has been shown only for isolated
acinar cells mimicking the early phase of pancreatitis. Tak-
ing into account pyroptosis and necroptosis, inammation is
the origin and consequence of cell death in pancreatitis
(Figure 3).
Protease Activity and Disease Severity
This raises the question of whether intracellular prote-
ase activation of trypsin as it is linked to cell death also can
mediate systemic disease severity. The notion that trypsin
activation is linked to disease severity is supported by the
correlation of TAP urine levels to severity in patients with
AP.
145
However, several studies have questioned the role of
trypsin for severity of pancreatitis. Expression of mutant
human trypsin bearing the HP mutation R122H in mice
leads to slightly more severe cerulein pancreatitis,
146
but,
when compared with mice expressing normal human tryp-
sinogen, there is no increase in disease severity. Moreover,
the effect seems to be not solely dependent on trypsin,
because mice with human trypsin mutations (R122H or
N29I) show lower trypsin activity after cerulein hyper-
stimulation. This might be explained in part by a higher rate
of acinar cell apoptosis even in untreated animals transgenic
for human trypsinogen.
147
A similar effect is seen in PACE-
tryp(on) mice, which conditionally express an endogenously
activated trypsinogen within pancreatic acinar cells. Those
mice will develop AP in a trypsin activity-dependent way,
which can lead to organ dysfunction and mortality, but they
also show pronounced caspase 3 activation with consecu-
tive apoptotic loss of acinar cells and replacement by fatty
tissue.
148
Similarly, Archer et al
149
described a transgenic
mouse, in which the human mutation R122H was inserted in
the murine PRSS1 gene. Those mice developed spontaneous
pancreatitis and showed a more pronounced inammatory
inltrate and cellular damage in response to cerulein. The
fact that the predominant way of cell death determines the
overall severity of experimental pancreatitis in mice has
been demonstrated in animals with deletion for cathepsin L.
Cathepsin L degrades trypsinogen into an inactive elongated
TAP, but cathepsin L deciency leads to a milder form a of
cerulein-induced pancreatitis, which is linked to a shift from
necrosis to apoptosis.
6
Themostconvincingdataquestioningtheroleof
trypsin for disease severity were generated using a mouse
strain lacking trypsinogen 7, the murine counterpart of
human cationic trypsinogen. Dawra et al
150
and Sah
et al
151
found that a signicant decrease in trypsin and
chymotrypsin activity after cerulein hyperstimulation in
these mice had no effect on disease severity for cell death
or local or systemic inammation in AP and CP models,
which they claimed was mediated by a mechanism
dependent on nuclear factor klight-chain enhancer of
activated B cells (NFkB). This conrms previous ndings
generatedinthePACE-tryp(on)model,inwhichNFkB
-activation was independent of increased intracellular
trypsin activity in vitro.
152
May 2019 Genetics and Pathophysiology of Pancreatitis 1957
PANCREATITIS
Figure 3. Trypsinogen activa-
tion and cell death in pancreatic
acinar cells. Intracellular tryp-
sinogen activation is an early
eventattheonsetofexperi-
mental AP. Supramaximal
secretagogue-receptor stimula-
tion or activation of bile salt re-
ceptors leads to an
unphysiologic peakplateau
calcium signal. This results in
disrupted exocytosis of
zymogen granules, secretory
blockage, zymogen retention,
and formation of EVs, which
contain trypsin and trypsin-
ogen, taken up from the extra-
cellular space. Those EVs
colocalize and fuse with lyso-
somes containing cathepsin B,
which in turn transforms tryp-
sinogen into active trypsin.
Owing to increasing instability,
EVs often rupture, releasing
trypsin and cathepsin B into the
cytosol. Active trypsin is
believed to induce mainly
apoptosis, a silent form of cell
death, which suppresses
inammation. In contrast, if the
pathologic calcium release
cannot be contained, then rapid
energy depletion occurs and
cells undergo necrosis, during
which the plasma membrane
becomes leaky and cellular
components (eg, DNA or mito-
chondria) reach the extracellular
space. Those will be recognized
by leukocytes, which will be
activated by the inammasome
signaling pathway. IL1band
TNFarelease and pyroptosis
occur. If TNFareaches the
basolateral membrane of previ-
ously unaffected or slightly
damaged acinar cells, then it
can induce another form of
programmed cell death (ie,
necroptosis). PLC, phospholi-
pase C.
1958 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
Role of Systemic Inammation in
Pancreatitis
NFkB ActivationInitial Step of Inammation
The activation of NFkB is an early event during
pancreatitis and occurs within the rst minutes after onset
of the disease.
153,154
One main function of NFkB is tran-
scriptional regulation of the immune response.
155
The
principal pathway of NFkB signal transduction is depicted in
Figure 4. The fact that NFkB is already present in the
cytoplasm explains its rapid activation after induction of
pancreatitis.
153,154
Intra-acinar protease activation and NFkB activation are
early cellular events during pancreatitis,
153,154
which have
been suggested to occur independent of each other,
151
but
follow a similar kinetic. Trypsinogen activation depends on
intracellular Ca
2þ
signaling
87
and NFkB activation also can
be induced by protein kinase C and Ca
2þ
, which could be the
reason for parallel kinetics.
156
The deletion of trypsinogen 7
or cathepsin B, either of which results in greatly decreased
protease activation,
104,144,151
does not inuence NFkB acti-
vation during cerulein-induced pancreatitis.
151
This can be
regarded as evidence that protease activation does not
directly lead to NFkB activation in acinar cells, whereas
NFkB activation can still interact with protease activity: the
transcriptional regulation of serine protease inhibitor 2a is
mediated by NFkB and can inhibit trypsin activity in a
mouse model of acute pancreatitis (Figure 4).
157
These re-
sults suggest a more complex role of NFkB during disease
progression that is not restricted to proinammation.
158
Surprisingly, the pancreas-specic deletion of Rel-A(p65)
resulted in increased systemic inammation and pancre-
atic necrosis.
159
In contrast, the pancreas-specic deletion
of NFkB inhibitor aresulted in nuclear translocation of Rel-
A and ameliorated pancreatitis.
157
The same phenotype was
observed in mice decient in myeloid differentiation pri-
mary response 88 (MyD88), which developed a more severe
disease phenotype compared with controls.
160
MyD88 is a
central adaptor for the TLRIL1-receptor signaling pathway,
which induces NFkB activation. These results ultimately
suggest a protective role of NFkB activation in pancreatic
acinar cells. Other studies attributed a more critical role for
NFkB in disease severity. Mice that constitutively over-
express the active inhibitor of NFkB kinase subunit b(IKKb)
under a pancreas-specic promoter showed chronic inl-
tration of immune cells associated with an increased disease
severity after cerulein stimulation.
161,162
However, the
inltration of immune cells alone, or the constitutive activity
of NFkB in acinar cells, did not result in AP, organ damage,
and necrotic or apoptotic cell death
162
; an additional path-
ophysiologic stimulus was still needed. The increased
severity of pancreatitis in IKKb-overexpressing mice can be
Figure 4. NFkBpathwayin
pancreatic acinar cells. The
early activation of NFkB
follows the same time
pattern as trypsinogen
activation. They are
induced by cytoplasmic
Ca
2þ
inux, but NFkBdoes
not depend on trypsinogen
activation. The phosphory-
lation of IkBa,followedby
proteosomal degradation
and nuclear translocation
of NFkB(p65p50) occurs
in parallel to protease acti-
vation. NFkB as a tran-
scription factor acts in 2
directions; rst, transcrip-
tions of proinammatory
genes, such as IL6 or
TNFa, initiate the immune
response; and second, the
transcription of prosurvival
genes. Therefore, NFkB
can directly inuence pro-
tease activity to protect
cells by the up-regulation
of serine protease inhibitor
2a. CTSB, cathepsin B;
IkBa, inhibitor of NFkB
alpha; IkBb, inhibitor of
NFkB beta; NEMO, NF-
kappa-B essential modu-
lator; PLC, phospholipase
C; Ub, ubiquitin.
May 2019 Genetics and Pathophysiology of Pancreatitis 1959
PANCREATITIS
explained by the presence of leukocytes within the
pancreas, which become rapidly activated after disease in-
duction and do not need to inltrate the pancreas. The
pancreas-specic deletion of IKKa, another NFkB inhibitor a
phosphorylating kinase, caused spontaneous pancreatitis in
mice,
163
but this process appeared to be independent of
NFkB. IKKaalso regulates autophagic ux, which is essential
for pancreas homeostasis.
164
These data demonstrate the
complexity of the NFkB network, which often hampers the
interpretation of results. Taken together, the constitutive
activation of NFkB leads to a chronic inltration of immune
cells, but pancreatitis develops only after induction by an
external stimulus.
161,162
The presence of immune cells
within the pancreas is required but insufcient for pancre-
atitis to develop and these cells need to be activated to
contribute to disease severity. Conversely, data from
pancreas-specic RelA deleted mice show that NFkB acti-
vation in acinar cells is not essential for the recruitment of
immune cells to the side of damage.
159
Quite to the contrary,
the absence of p65 in acinar cells result in greater disease
severity. Although all these studies investigated the role of
NFkB in acinar cells, NFkB activation could play a much
larger role in inltrating immune cells, which directly
regulate the immune response.
Another master switch in the transcriptional machinery
of acinar cells is activator protein 1 (AP1). AP1 is implicated
in multiple transcriptional networks within the acinar cells
regulating pancreatic development, differentiation, cell
death, and inammation. Mice heterozygous for the orphan
nuclear receptor NR5A2 develop an acinar cellautonomous
AP1-dependent pre-inammatory state, which, on a tran-
scriptome level, mimics that of early AP.
165
However, the extent of NFkB and AP1 activity seems to
greatly differ with respect to the cause of pancreatitis. In
cerulein models, activation of the 2 transcription factors is
described and submaximal CCK stimulation induces acinar
cell dedifferentiation and proliferation through the mitogen-
activated kinasec-JunAP1 pathway probably as part of a
pancreatic regeneration program.
166,167
In contrast, the
metabolites occurring in ethanol-induced experimental
pancreatitis can positively and negatively regulate NFkB and
AP1 depending on the predominance of oxidative or non-
oxidative alcohol metabolism in the pancreas.
168
Role of Inltrating Immune Cells
Inltration of immune cells starts within minutes after the
onset of disease and plays a crucial role in the severity and
prognosis of pancreatitis.
169172
Cells of the innate immune
system, such as neutrophil granulocytes and monocytes and
macrophages, represent the majority of inltrating cells.
NFkB plays a crucial role in the activation of leukocytes and is
a central mediator of the innate and adaptive immune sys-
tem.
173
Pancreatitis is primarily a sterile inammation, so
pathogen-associated molecular patterns play no role in the
activation of immune cells during the early phase of disease.
Thus, in pancreatitis, activation of immune cells is mediated
by cytokines or DAMPs that arise from acinar cell necrosis.
Acinar cells release various cytokines and chemokines in
response to CCK stimulation and NFkB activation, such as
TNFa,
174
IL6, or monocyte chemoattractant protein 1.
175
DAMPs and cytokines result in the nuclear translocation of
p65 and p50 within inltrating immune cells, which enhances
the cytokine storm through the secretion of proinammatory
mediators
175
(Figure 5). DAMPs can act in the same way as
pathogen-associated molecular patterns through TLRs
176
or
specic receptors such as P2RX7, which uses extracellular
adenosine triphosphate as a ligand. Acinar cells, which un-
dergo necrotic cell death, release a multitude of different
DAMPs, such as free DNA,
177
histones, or free adenosine
triphosphate,
138
which can act as immune activators. Acti-
vated immune cells increase pancreatic damage and
contribute to systemic inammation.
169,175,178
Several studies
have focused on different populations of immune cells and
their role during AP.
Neutrophil granulocytes are often used as reference
markers for pancreatic inammation by measurements of
myeloperoxidase activity in tissue and reect the amount of
inltrating neutrophils. One major function of neutrophils is
removing pathogens by the release of proteases, antimi-
crobial peptides, and ROS. During pancreatitis, neutrophils
are the major source of ROS production; they can induce
oxidative damage on acinar cells and enhance trypsinogen
activation.
178
The release of proteases such as PMN-elastase
contributes to tissue destruction and acinar cell dissocia-
tion.
179
These data indicate that neutrophils have a direct
effect on disease severity. This was conrmed by the
depletion of neutrophils using antineutrophil serum, which
resulted in decreased pancreatic damage and protease
activation.
169,178
Recent studies have investigated the role of
neutrophil extracellular trap (NET) formation in the context
of pancreatitis. NETs are extracellular networks consisting
of neutrophil DNA and are used to bind pathogens.
180
This
suicide mechanism of neutrophils is a last line of defense
against bacterial infections. NET formation is induced by
TLR4 activation and the activation of nicotinamide adenine
dinucleotide phosphate oxidase, which lead to the oxidation
of peptidyl-arginine-deiminase 4.
181,182
TLR4 is responsible
for the detection of pathogens, although DAMPs can activate
the TLR-signaling pathway.
176
Merza et al
183
reported that
NET formation enhances the immune response during se-
vere AP and is accountable for trypsinogen activation.
Treatment with DNAse prevents NET formation and de-
creases disease severity.
183
In addition to bacterial in-
fections, crystals can induce NET formation.
184
Another
group showed that NET formation is a critical step in bile
stone development and plays an important role in ductal
obstruction contributing to onset and severity of pancrea-
titis.
185,186
Neutrophil inltration during AP is an unspecic
reaction of the immune system, which enhances local
damage by the formation of NETs and the release of ROS or
activates digestive enzymes.
Monocytes and macrophages belong to cells of the innate
immune system. In contrast to neutrophil granulocytes,
macrophages are characterized by high plasticity. Classic
activated macrophages (M1) act in a proinammatory
manner and secrete large amounts of IL6, TNFa, IL12, and
IL1b, and increased expression of inducible nitric oxide
1960 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
synthase results in the release of nitric oxide.
187
Alterna-
tively, activated macrophages (M2) are associated with
wound healing, tissue regeneration, and brogenesis and act
in an anti-inammatory manner through the release of IL10
or transforming growth factor b. They are characterized by
decreased inducible nitric oxide synthase and increased argi-
nase 1 expression.
187
Macrophages are phagocytosing cells that
remove tissue debris and necrotic and apoptotic cells. During
AP, macrophage inltration correlates to a greater extent with
pancreatic damage and necrosis than with the number of
Figure 5. NFkB activation in inammatory cells. There are multiple pathways of how NFkB could be activated within leuko-
cytes; the major pathways that play a role during AP are depicted. Cytokines such as IL1bor TNFaand DAMP signals acting
by TLRs can induce the translocation of p65 and p50 into the nucleus. In leukocytes, most inammatory mediators are under
the control of NFkB: cytokines, chemokines, adhesion molecules, and components of the inammasome pathway. Leukocyte-
mediated NFkB activation enhances the immune response in a very prominent manner and therefore has a different role in
pancreatitis compared with NFkB activation within the acinar cell. ASC, Apoptosis-associated speck-like protein containing a
CARD; Casp-1, caspase 1; ICAM-1, intercellular adhesion molecule 1; IL1-R, IL1 receptor; IRAK, interleukin-1 receptor-
associated kinase; IkBa, inhibitor of NFkB alpha; MCP-1, monocyte chemoattractant protein 1; MIP-2, macrophage inam-
matory protein 2; MMPs, matrix metalloproteinases; NEMO, NF-kappa-B essential modulator; RANTES, regulated on acti-
vation, normal T-cell expressed, and secreted; TAK1, transforming growth factor beta-activated kinase 1; TNFR, TNF receptor;
TRADD, tumor necrosis factor receptor type 1-associated DEATH domain protein; TRAF2, TNF receptor-associated factor 2;
Ub, ubiquitin; VCAM-1, vascular adhesion molecule 1.
May 2019 Genetics and Pathophysiology of Pancreatitis 1961
PANCREATITIS
inltrating neutrophils.
175
The reason for that is that macro-
phages are required for the removal of necrosis and thus miti-
gate pancreatic damage. Phagocytosing macrophages have been
observed in different models of AP and CP.
130,164,169,175
In
contrast to apoptosis, necrosis is a proinammatory cell death
because it entails the release of multiple DAMPs that induce an
M1 polarization of macrophages.
188
Therefore, acinar cell
apoptosis is suggested to be protective against hyper-
inammation and decrease disease severity.
189
M1 macro-
phages release large amounts of TNFa, which have a direct
effect on pancreatic acinar cells.
174
Two independent groups
reported that TNFasecreted from inltrating monocytes is
responsible for pancreatic damage and digestive protease
activation.
169,190
Depletion of macrophages by clodronate-
containing liposomes decreased disease severity and pro-
tected mice from cerulein-induced pancreatitis.
169,191
TNFa
acts on cells by cell death receptors and is necessary to induce
cell death by necroptosis through the RIP1RIP3 pathway,
135
which has been suggested to be the major cell death pathway
during AP.
136
Therefore, inltrating macrophages are respon-
sible for the induction of necroptosis and for the clearance of
necrotic areas within the damaged pancreas.
175
In addition to TNFa, macrophages produce large amounts
of IL1b, a proinammatory cytokine associated with the
acute disease phase. In contrast to other cytokines, IL1b
needs to be processed. During maturation, pro-IL1band pro-
IL18 undergo activation by the caspase 1inammasome
complex and are released by the gasdermin Dpore complex
from the cytosol to the extracellular space.
192
In consequence
of this process, the cell undergoes pyroptotic cell death.
192,193
During pancreatitis, the activation of the inammasome
complex and the release of IL1bcontribute critically to dis-
ease severity.
138,139,175,177
Inammasome complex formation
and pyroptotic cell death are mainly known from macro-
phages and are not present in acinar cells.
175
Inammasome
activation is a complex mechanism requiring 2 signals for
activation: the rst signal induces transcriptional up-
regulation of inammasome components by NFkB and the
second signal induces oligomerization of the inammasome
complex and the activation of procaspase 1. The major
inducer of the inammasome pathway is the TLRMyD88
cascade. The second signal, which is necessary for inam-
masome complex formation, can be a high potassium inux,
TNFa, or the release of cathepsins from phagosomes into the
cytosol.
194
Phagocytosis of zymogens by macrophages results
in a colocalization of trypsinogen and cathepsin B in non
acinar cell phagolysosomes and results in activation of tryp-
sinogen
175
and macrophage activation. The rupture of these
trypsin- and cathepsin Bcontaining vesicles leads to a
cytosolic redistribution of cathepsins that acts as a second
signal of inammasome activation and indirectly links tryp-
sinogen activation to the NFkB pathway through IL1brelease.
IL1bacts as an activator for other immune cells, with the IL1
receptor being directly linked to the MyD88NFkB pathway.
The importance of IL1bduring pancreatitis was nicely shown
by a transgenic mouse model of pancreas-specicIL1b-
overexpressing mice that developed CP, including complete
loss of pancreatic function within weeks after birth.
195
Therefore, M1 macrophages contribute prominently to the
systemic immune response syndrome, which is associated
with multiorgan dysfunction syndrome and increased mor-
tality.
170
In contrast to M1 macrophages, the M2 phenotype is
associated with organ regeneration and brosis devel-
opment.
196198
In conclusion, early protease activation and NFkB acti-
vation are essential characteristics of pancreatitis; these 2
events occur in parallel during disease manifestation and
strongly inuence each other. Recent studies have proved
that not only the activation of proteases and NFkB but also
the type of cell play a critical role, in which where their
activation takes place is of importance. Pancreatitis is not a
disease of acinar cells alone.
Supplementary Material
Note: To access the supplementary material accompanying
this article, visit the online version of Gastroenterology at
www.gastrojournal.org, and at https://doi.org/10.1053/j.
gastro.2018.11.081.
References
1. Peery AF, Crockett SD, Murphy CC, et al. Burden and
cost of gastrointestinal, liver, and pancreatic diseases in
the United States: update 2018. Gastroenterology 2019;
156:254272.e11.
2. Yadav D, Lowenfels AB. The epidemiology of pancrea-
titis and pancreatic cancer. Gastroenterology 2013;
144:12521261.
3. Rinderknecht H. Activation of pancreatic zymogens.
Normal activation, premature intrapancreatic activation,
protective mechanisms against inappropriate activation.
Dig Dis Sci 1986;31:314321.
4. Szmola R, Bence M, Carpentieri A, et al. Chymotrypsin C
is a co-activator of human pancreatic procarbox-
ypeptidases A1 and A2. J Biol Chem 2011;286:1819
1827.
5. Szmola R, Sahin-Tóth M. Chymotrypsin C (caldecrin)
promotes degradation of human cationic trypsin: identity
with Rinderknechts enzyme Y. Proc Natl Acad Sci U S A
2007;104:1122711232.
6. Wartmann T, Mayerle J, Kähne T, et al. Cathepsin L
inactivates human trypsinogen, whereas cathepsin
L-deletion reduces the severity of pancreatitis in mice.
Gastroenterology 2010;138:726737.
7. Hegyi E, Sahin-Tóth M. Genetic risk in chronic pancre-
atitis: the trypsin-dependent pathway. Dig Dis Sci 2017;
62:16921701.
8. Nemoda Z, Sahin-Tóth M. Chymotrypsin C (caldecrin)
stimulates autoactivation of human cationic trypsinogen.
J Biol Chem 2006;281:1187911886.
9. Szabó A, Sahin-Tóth M. Increased activation of heredi-
tary pancreatitis-associated human cationic trypsinogen
mutants in presence of chymotrypsin C. J Biol Chem
2012;287:2070120710.
10. Weiss FU, Behn C-O, Simon P, et al. Cathepsin B gene
polymorphism Val26 is not associated with idiopathic
chronic pancreatitis in European patients. Gut 2007;
56:13221323.
1962 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
11. Németh BC, Sahin-Tóth M. Human cationic trypsinogen
(PRSS1) variants and chronic pancreatitis. Am J Physiol
Gastrointest Liver Physiol 2014;306:G466G473.
12. Németh BC, Szücs Á, Hegyi P, et al. Novel PRSS1
mutation p.P17T validates pathogenic relevance of
CTRC-mediated processing of the trypsinogen activa-
tion peptide in chronic pancreatitis. Am J Gastroenterol
2017;112:18961898.
13. Chen J-M, Kukor Z, Le Maréchal C, et al. Evolution of
trypsinogen activation peptides. Mol Biol Evol 2003;
20:17671777.
14. Geisz A, Hegyi P, Sahin-Tóth M. Robust autoactivation,
chymotrypsin C independence and diminished secretion
dene a subset of hereditary pancreatitis-associated
cationic trypsinogen mutants. FEBS J 2013;280:2888
2899.
15. Joergensen MT, Geisz A, Brusgaard K, et al. Intragenic
duplication: a novel mutational mechanism in hereditary
pancreatitis. Pancreas 2011;40:540546.
16. Kereszturi E, Sahin-Tóth M. Intracellular autoactivation
of human cationic trypsinogen mutants causes reduced
trypsinogen secretion and acinar cell death. J Biol Chem
2009;284:3339233399.
17. Whitcomb DC, LaRusch J, Krasinskas AM, et al. Com-
mon genetic variants in the CLDN2 and PRSS1-PRSS2
loci alter risk for alcohol-related and sporadic pancrea-
titis. Nat Genet 2012;44:13491354.
18. DerikxMH,KovacsP,ScholzM,etal.Polymorphisms
at PRSS1-PRSS2 and CLDN2-MORC4 loci associate
with alcoholic and non-alcoholic chronic pancreatitis
in a European replication study. Gut 2015;64:1426
1433.
19. Rosendahl J, Kirsten H, Hegyi E, et al. Genome-wide
association study identies inversion in the CTRB1-
CTRB2 locus to modify risk for alcoholic and non-
alcoholic chronic pancreatitis. Gut 2018;67:18551863.
20. Boulling A, Sato M, Masson E, et al. Identication of a
functional PRSS1 promoter variant in linkage disequi-
librium with the chronic pancreatitis-protecting
rs10273639. Gut 2015;64:18371838.
21. Witt H, Luck W, Hennies HC, et al. Mutations in the gene
encodingtheserineproteaseinhibitor,Kazaltype1are
associated with chronic pancreatitis. Nat Genet 2000;
25:213216.
22. Aoun E, Chang C-CH, Greer JB, et al. Pathways to injury
in chronic pancreatitis: decoding the role of the high-risk
SPINK1 N34S haplotype using meta-analysis. PloS One
2008;3:e2003.
23. Di Leo M, Bianco M, Zuppardo RA, et al. Meta-analysis
of the impact of SPINK1 p.N34S gene variation in Cau-
casic patients with chronic pancreatitis. An update. Dig
Liver Dis 2017;49:847853.
24. Kuwata K, Hirota M, Shimizu H, et al. Functional analysis
of recombinant pancreatic secretory trypsin inhibitor
protein with amino-acid substitution. J Gastroenterol
2002;37:928934.
25. Király O, Wartmann T, Sahin-Tóth M. Missense muta-
tions in pancreatic secretory trypsin inhibitor (SPINK1)
cause intracellular retention and degradation. Gut 2007;
56:14331438.
26. Boulling A, Le Maréchal C, Trouvé P, et al. Functional
analysis of pancreatitis-associated missense mutations
in the pancreatic secretory trypsin inhibitor (SPINK1)
gene. Eur J Hum Genet 2007;15:936942.
27. Kereszturi E, Király O, Sahin-Tóth M. Minigene analysis of
intronic variants in common SPINK1 haplotypes associ-
ated with chronic pancreatitis. Gut 2009;58:545549.
28. Kereszturi É, Sahin-Tóth M. Pancreatic cancer cell lines
heterozygous for the SPINK1 p.N34S haplotype exhibit
diminished expression of the variant allele. Pancreas
2017;46:e54e55.
29. Pfützer RH, Barmada MM, Brunskill AP, et al. SPINK1/
PSTI polymorphisms act as disease modiers in familial
and idiopathic chronic pancreatitis. Gastroenterology
2000;119:615623.
30. Kume K, Masamune A, Kikuta K, et al. [215G>A;
IVS3þ2T>C] mutation in the SPINK1 gene causes exon
3 skipping and loss of the trypsin binding site. Gut 2006;
55:1214.
31. Zou W-B, Boulling A, Masson E, et al. Clarifying the
clinical relevance of SPINK1 intronic variants in chronic
pancreatitis. Gut 2016;65:884886.
32. Boulling A, Witt H, Chandak GR, et al. Assessing the
pathological relevance of SPINK1 promoter variants. Eur
J Hum Genet 2011;19:10661073.
33. Derikx MHM, Geisz A, Kereszturi É, et al. Functional
signicance of SPINK1 promoter variants in chronic
pancreatitis. Am J Physiol Gastrointest Liver Physiol
2015;308:G779G784.
34. Kukor Z, Tóth M, Sahin-Tóth M. Human anionic tryp-
sinogen: properties of autocatalytic activation and
degradation and implications in pancreatic diseases. Eur
J Biochem 2003;270:20472058.
35. Witt H, Sahin-Tóth M, Landt O, et al. A degradation-
sensitive anionic trypsinogen (PRSS2) variant protects
against chronic pancreatitis. Nat Genet 2006;38:668673.
36. Jancsó Z, Sahin-Tóth M. Tighter control by chymo-
trypsin C (CTRC) explains lack of association between
human anionic trypsinogen and hereditary pancreatitis.
J Biol Chem 2016;291:1289712905.
37. Kume K, Masamune A, Takagi Y, et al. A loss-of-function
p.G191R variant in the anionic trypsinogen (PRSS2)
gene in Japanese patients with pancreatic disorders. Gut
2009;58:820824.
38. Rosendahl J, Witt H, Szmola R, et al. Chymotrypsin C
(CTRC) variants that diminish activity or secretion are
associated with chronic pancreatitis. Nat Genet 2008;
40:7882.
39. Rosendahl J, Landt O, Bernadova J, et al. CFTR,
SPINK1, CTRC and PRSS1 variants in chronic pancre-
atitis: is the role of mutated CFTR overestimated? Gut
2013;62:582592.
40. Beer S, Zhou J, Szabó A, et al. Comprehensive func-
tional analysis of chymotrypsin C (CTRC) variants re-
veals distinct loss-of-function mechanisms associated
with pancreatitis risk. Gut 2013;62:16161624.
41. Szabó A, Ludwig M, Hegyi E, et al. Mesotrypsin signa-
ture mutation in a chymotrypsin C (CTRC) variant
associated with chronic pancreatitis. J Biol Chem 2015;
290:1728217292.
May 2019 Genetics and Pathophysiology of Pancreatitis 1963
PANCREATITIS
42. Masson E, Chen J-M, Scotet V, et al. Association of rare
chymotrypsinogen C (CTRC) gene variations in patients
with idiopathic chronic pancreatitis. Hum Genet 2008;
123:8391.
43. Paliwal S, Bhaskar S, Mani KR, et al. Comprehensive
screening of chymotrypsin C (CTRC) gene in tropical
calcic pancreatitis identies novel variants. Gut 2013;
62:16021606.
44. LaRusch J, Lozano-Leon A, Stello K, et al. The common
chymotrypsinogen C (CTRC) variant G60G (C.180T) in-
creases risk of chronic pancreatitis but not recurrent
acute pancreatitis in a North American population. Clin
Transl Gastroenterol 2015;6:e68.
45. Grabarczyk AM, Oracz G, Wertheim-Tysarowska K, et al.
Chymotrypsinogen C genetic variants, including c.
180TT, are strongly associated with chronic pancreatitis
in pediatric patients. J Pediatr Gastroenterol Nutr 2017;
65:652657.
46. Tang X-Y, Zou W-B, Masson E, et al. The CTRB1-CTRB2
risk allele for chronic pancreatitis discovered in Euro-
pean populations does not contribute to disease risk
variation in the Chinese population due to near allele
xation. Gut 2018;67:13681369.
47. Jancsó Z, Hegyi E, Sahin-Tóth M. Chymotrypsin reduces
the severity of secretagogue-induced pancreatitis in
mice. Gastroenterology 2018;155:10171021.
48. Sahin-Tóth M. Genetic risk in chronic pancreatitis: the
misfolding-dependent pathway. Curr Opin Gastroenterol
2017;33:390395.
49. Kereszturi E, Szmola R, Kukor Z, et al. Hereditary
pancreatitis caused by mutation-induced misfolding of
human cationic trypsinogen: a novel disease mecha-
nism. Hum Mutat 2009;30:575582.
50. Masamune A, Nakano E, Kume K, et al. PRSS1 c.
623G>C (p.G208A) variant is associated with pancrea-
titis in Japan. Gut 2014;63:366.
51. Hegyi E, Cierna I, Vavrova L, et al. Chronic pancreatitis
associated with the p.G208A variant of PRSS1 gene in a
European patient. J Pancreas 2014;15:4952.
52. Witt H, Beer S, Rosendahl J, et al. Variants in CPA1 are
strongly associated with early onset chronic pancreatitis.
Nat Genet 2013;45:12161220.
53. Kujko AA, Berki DM, Oracz G, et al. A novel p.Ser282Pro
CPA1 variant is associated with autosomal dominant
hereditary pancreatitis. Gut 2017;66:17281730.
54. Nakano E, Geisz A, Masamune A, et al. Variants in
pancreatic carboxypeptidase genes CPA2 and CPB1 are
not associated with chronic pancreatitis. Am J Physiol
Gastrointest Liver Physiol 2015;309:G688G694.
55. Tamura K, Yu J, Hata T, et al. Mutations in the pancreatic
secretory enzymes CPA1 and CPB1 are associated with
pancreatic cancer. Proc Natl Acad Sci U S A 2018;
115:47674772.
56. Hegyi E, Sahin-Tóth M. Human CPA1 mutation causes
digestive enzyme misfolding and chronic pancreatitis in
mice. Gut 2019;68:301312.
57. Raeder H, Johansson S, Holm PI, et al. Mutations in
the CEL VNTR cause a syndrome of diabetes and
pancreatic exocrine dysfunction. Nat Genet 2006;
38:5462.
58. Johansson BB, Torsvik J, Bjørkhaug L, et al. Diabetes
and pancreatic exocrine dysfunction due to mutations in
the carboxyl ester lipase gene-maturity onset diabetes of
the young (CEL-MODY): a protein misfolding disease.
J Biol Chem 2011;286:3459334605.
59. Xiao X, Jones G, Sevilla WA, et al. A carboxyl ester lipase
(CEL) mutant causes chronic pancreatitis by forming
intracellular aggregates that activate apoptosis. J Biol
Chem 2016;291:2322423236.
60. Fjeld K, Weiss FU, Lasher D, et al. A recombined allele of
the lipase gene CEL and its pseudogene CELP confers
susceptibility to chronic pancreatitis. Nat Genet 2015;
47:518522.
61. Zou W-B, Boulling A, Masamune A, et al. No association
between CEL-HYB hybrid allele and chronic pancreatitis
in Asian populations. Gastroenterology 2016;150:1558
1560.e5.
62. Weiss FU, Schurmann C, Guenther A, et al. Fucosyl-
transferase 2 (FUT2) non-secretor status and blood
group B are associated with elevated serum lipase ac-
tivity in asymptomatic subjects, and an increased risk for
chronic pancreatitis: a genetic association study. Gut
2015;64:646656.
63. Kirsten H, Scholz M, Kovacs P, et al. Genetic variants of
lipase activity in chronic pancreatitis. Gut 2016;65:184185.
64. Greer JB, LaRusch J, Brand RE, et al. ABO blood group
and chronic pancreatitis risk in the NAPS2 cohort.
Pancreas 2011;40:11881194.
65. Teich N, Bokemeyer B, Mohl W, et al. Blood group B is
associated with azathioprine-induced acute pancreatitis
in patients with IBD. Gut 2017;66:15311532.
66. Hegyi P, Wilschanski M, Muallem S, et al. CFTR: a new
horizon in the pathomechanism and treatment of pancre-
atitis. Rev Physiol Biochem Pharmacol 2016;170:3766.
67. Sharer N, Schwarz M, Malone G, et al. Mutations of the
cystic brosis gene in patients with chronic pancreatitis.
N Engl J Med 1998;339:645652.
68. Cohn JA, Friedman KJ, Noone PG, et al. Relation be-
tween mutations of the cystic brosis gene and idio-
pathic pancreatitis. N Engl J Med 1998;339:653658.
69. Weiss FU, Simon P, Bogdanova N, et al. Complete
cystic brosis transmembrane conductance regulator
gene sequencing in patients with idiopathic chronic
pancreatitis and controls. Gut 2005;54:14561460.
70. Masson E, Chen J-M, Audrézet M-P, et al.
A conservative assessment of the major genetic causes
of idiopathic chronic pancreatitis: data from a compre-
hensive analysis of PRSS1, SPINK1, CTRC and CFTR
genes in 253 young French patients. PloS One 2013;
8:e73522.
71. LaRusch J, Jung J, General IJ, et al. Mechanisms of
CFTR functional variants that impair regulated bicar-
bonate permeation and increase risk for pancreatitis but
not for cystic brosis. PLoS Genet 2014;10:e1004376.
72. Balázs A, Ruffert C, Hegyi E, et al. Genetic analysis of the
bicarbonate secreting anion exchanger SLC26A6 in
chronic pancreatitis. Pancreatology 2015;15:508513.
73. Rácz GZ, Kittel A, Riccardi D, et al. Extracellular calcium
sensing receptor in human pancreatic cells. Gut 2002;
51:705711.
1964 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
74. Muddana V, Lamb J, Greer J-B, et al. Association be-
tween calcium sensing receptor gene polymorphisms
and chronic pancreatitis in a US population: role of
serine protease inhibitor Kazal 1type and alcohol. World
J Gastroenterol 2008;14:44864491.
75. Masson E, Chen J-M, Férec C. Overrepresentation of
rare CASR coding variants in a sample of young French
patients with idiopathic chronic pancreatitis. Pancreas
2015;44:996998.
76. Chiari H. Über die Selbstverdauung des menschlichen
Pankreas. Z Heilkd 1896:6996.
77. Bialek R, Willemer S, Arnold R, et al. Evidence of intra-
cellular activation of serine proteases in acute cerulein-
induced pancreatitis in rats. Scand J Gastroenterol
1991;26:190196.
78. Hofbauer B, Saluja AK, Lerch MM, et al. Intra-acinar cell
activation of trypsinogen during caerulein-induced
pancreatitis in rats. Am J Physiol Gastrointest Liver
Physiol 1998;275:G352G362.
79. Fernández-del Castillo C, Harringer W, Warshaw AL,
et al. Risk factors for pancreatic cellular injury after car-
diopulmonary bypass. N Engl J Med 1991;325:382387.
80. Mithöfer K, Fernández-del Castillo C, Frick TW, et al.
Acute hypercalcemia causes acute pancreatitis and
ectopic trypsinogen activation in the rat. Gastroenter-
ology 1995;109:239246.
81. Bai HX, Giefer M, Patel M, et al. The association of pri-
mary hyperparathyroidism with pancreatitis. J Clin
Gastroenterol 2012;46:656661.
82. Kasai H, Augustine GJ. Cytosolic Ca2þgradients trig-
gering unidirectional uid secretion from exocrine
pancreas. Nature 1990;348:735738.
83. Gerasimenko JV, Gerasimenko OV, Petersen OH. The
role of Ca2þin the pathophysiology of pancreatitis.
J Physiol 2014;592:269280.
84. Lampel M, Kern HF. Acute interstitial pancreatitis inthe rat
induced by excessive doses of a pancreatic secretagogue.
Virchows Arch A Pathol Anat Histol 1977;373:97117.
85. Ward JB, Sutton R, Jenkins SA, et al. Progressive
disruption of acinar cell calcium signaling is an early
feature of cerulein-induced pancreatitis in mice.
Gastroenterology 1996;111:481491.
86. Krüger B, Lerch MM, Tessenow W. Direct detection of
premature protease activation in living pancreatic acinar
cells. Lab Investig J Tech Methods Pathol 1998;78:763
764.
87. Krüger B, Albrecht E, Lerch MM. The role of intracellular
calcium signaling in premature protease activation and
the onset of pancreatitis. Am J Pathol 2000;157:4350.
88. Raraty M, Ward J, Erdemli G, et al. Calcium-dependent
enzyme activation and vacuole formation in the apical
granular region of pancreatic acinar cells. Proc Natl Acad
Sci U S A 2000;97:1312613131.
89. Mooren FC, Hlouschek V, Finkes T, et al. Early changes
in pancreatic acinar cell calcium signaling after pancre-
atic duct obstruction. J Biol Chem 2003;278:93619369.
90. Schick V, Scheiber JA, Mooren FC, et al. Effect of
magnesium supplementation and depletion on the onset
and course of acute experimental pancreatitis. Gut 2014;
63:14691480.
91. Orabi AI, Shah AU, Muili K, et al. Ethanol enhances
carbachol-induced protease activation and accelerates
Ca2þwaves in isolated rat pancreatic acini. J Biol Chem
2011;286:1409014097.
92. Muili KA, Wang D, Orabi AI, et al. Bile acids induce
pancreatic acinar cell injury and pancreatitis by acti-
vating calcineurin. J Biol Chem 2013;288:570580.
93. Romac JM-J, Shahid RA, Swain SM, et al. Piezo1 is a
mechanically activated ion channel and mediates pres-
sure induced pancreatitis. Nat Commun 2018;9:1715.
94. Gerasimenko JV, Lur G, Sherwood MW, et al. Pancreatic
protease activation by alcohol metabolite depends on
Ca2þrelease via acid store IP3 receptors. Proc Natl
Acad Sci U S A 2009;106:1075810763.
95. Huang W, Cane MC, Mukherjee R, et al. Caffeine pro-
tects against experimental acute pancreatitis by inhibi-
tion of inositol 1,4,5-trisphosphate receptor-mediated
Ca2þrelease. Gut 2017;66:301313.
96. Lur G, Sherwood MW, Ebisui E, et al. InsPreceptors and
Orai channels in pancreatic acinar cells: co-localization
and its consequences. Biochem J 2011;436:231239.
97. Wen L, Voronina S, Javed MA, et al. Inhibitors of ORAI1
prevent cytosolic calcium-associated injury of human
pancreatic acinar cells and acute pancreatitis in 3 mouse
models. Gastroenterology 2015;149:481492.e7.
98. Husain SZ, Grant WM, Gorelick FS, et al. Caerulein-
induced intracellular pancreatic zymogen activation is
dependent on calcineurin. Am J Physiol Gastrointest
Liver Physiol 2007;292:G1594G1599.
99. Muili KA, Ahmad M, Orabi AI, et al. Pharmacological and
genetic inhibition of calcineurin protects against
carbachol-induced pathological zymogen activation and
acinar cell injury. Am J Physiol Gastrointest Liver Physiol
2012;302:G898G905.
100. Kassell B, Kay J. Zymogens of proteolytic enzymes.
Science 1973;180:10221027.
101. Halangk W, Krüger B, Ruthenbürger M, et al. Trypsin
activity is not involved in premature, intrapancreatic
trypsinogen activation. Am J Physiol Gastrointest Liver
Physiol 2002;282:G367G374.
102. Saluja AK, Donovan EA, Yamanaka K, et al. Cerulein-
induced in vitro activation of trypsinogen in rat pancre-
atic acini is mediated by cathepsin B. Gastroenterology
1997;113:304310.
103. Van Acker GJD, Saluja AK, Bhagat L, et al. Cathepsin B
inhibition prevents trypsinogen activation and reduces
pancreatitis severity. Am J Physiol Gastrointest Liver
Physiol 2002;283:G794G800.
104. Halangk W, Lerch MM, Brandt-Nedelev B, et al. Role of
cathepsin B in intracellular trypsinogen activation and the
onset of acute pancreatitis. J Clin Invest 2000;106:773781.
105. Kukor Z, Mayerle J, Krüger B, et al. Presence of
cathepsin B in the human pancreatic secretory pathway
and its role in trypsinogen activation during hereditary
pancreatitis. J Biol Chem 2002;277:2138921396.
106. Talukdar R, Sareen A, Zhu H, et al. Release of cathepsin
B in cytosol causes cell death in acute pancreatitis.
Gastroenterology 2016;151:747758.e5.
107. Koike H, Steer ML, Meldolesi J. Pancreatic effects of
ethionine: blockade of exocytosis and appearance of
May 2019 Genetics and Pathophysiology of Pancreatitis 1965
PANCREATITIS
crinophagy and autophagy precede cellular necrosis.
Am J Physiol Gastrointest Liver Physiol 1982;242:G297
G307.
108. Saito I, Hashimoto S, Saluja A, et al. Intracellular trans-
port of pancreatic zymogens during caerulein supra-
maximal stimulation. Am J Physiol Gastrointest Liver
Physiol 1987;253:G517G526.
109. Saluja A, Hashimoto S, Saluja M, et al. Subcellular
redistribution of lysosomal enzymes during caerulein-
induced pancreatitis. Am J Physiol Gastrointest Liver
Physiol 1987;253:G508G516.
110. Saluja A, Saluja M, Villa A, et al. Pancreatic duct
obstruction in rabbits causes digestive zymogen and
lysosomal enzyme colocalization. J Clin Invest 1989;
84:12601266.
111. Hirano T, Saluja A, Ramarao P, et al. Apical secretion of
lysosomal enzymes in rabbit pancreas occurs via a
secretagogue regulated pathway and is increased after
pancreatic duct obstruction. J Clin Invest 1991;87:865
869.
112. Lerch MM, Saluja AK, Rünzi M, et al. Luminal endocy-
tosis and intracellular targeting by acinar cells during
early biliary pancreatitis in the opossum. J Clin Invest
1995;95:22222231.
113. Thrower EC, Diaz de Villalvilla APE, Kolodecik TR, et al.
Zymogen activation in a reconstituted pancreatic acinar
cell system. Am J Physiol Gastrointest Liver Physiol
2006;290:G894G902.
114. Bhoomagoud M, Jung T, Atladottir J, et al. Reducing
extracellular pH sensitizes the acinar cell to
secretagogue-induced pancreatitis responses in rats.
Gastroenterology 2009;137:10831092.
115. Dolai S, Liang T, Orabi AI, et al. Pancreatitis-induced
depletion of syntaxin 2 promotes autophagy and in-
creases basolateral exocytosis. Gastroenterology 2018;
154:18051821.e5.
116. Dolai S, Liang T, Orabi AI, et al. Depletion of the
membrane-fusion regulator Munc18c attenuates caer-
ulein hyperstimulation-induced pancreatitis. J Biol Chem
2018;293:25102522.
117. Behrendorff N, Dolai S, Hong W, et al. Vesicle-associ-
ated membrane protein 8 (VAMP8) is a SNARE (soluble
N-ethylmaleimidesensitive factor attachment protein
receptor) selectively required for sequential granule-to-
granule fusion. J Biol Chem 2011;286:2962729634.
118. Voronina S, Collier D, Chvanov M, et al. The role of Ca2þ
inux in endocytic vacuole formation in pancreatic acinar
cells. Biochem J 2015;465:405412.
119. Sherwood MW, Prior IA, Voronina SG, et al. Activation of
trypsinogen in large endocytic vacuoles of pancreatic
acinar cells. Proc Natl Acad Sci U S A 2007;104:5674
5679.
120. Chvanov M, De Faveri F, Moore D, et al. Intracellular
rupture, exocytosis and actin interaction of endocytic
vacuoles in pancreatic acinar cells: initiating events in
acute pancreatitis. J Physiol 2018;596:25472564.
121. Messenger SW, Thomas DD, Cooley MM, et al. Early to
late endosome trafcking controls secretion and zymogen
activation in rodent and human pancreatic acinar cells. Cell
Mol Gastroenterol Hepatol 2015;1:695709.
122. Messenger SW, Jones EK, Holthaus CL, et al. Acute
acinar pancreatitis blocks vesicle-associated membrane
protein 8 (VAMP8)-dependent secretion, resulting in
intracellular trypsin accumulation. J Biol Chem 2017;
292:78287839.
123. Thomas DDH, Martin CL, Weng N, et al. Tumor protein
D52 expression and Ca2þ-dependent phosphorylation
modulates lysosomal membrane protein trafcking to
the plasma membrane. Am J Physiol Cell Physiol 2010;
298:C725C739.
124. Messenger SW, Falkowski MA, Groblewski GE. Ca
2þ
-
regulated secretory granule exocytosis in pancreatic and
parotid acinar cells. Cell Calcium 2014;55:369375.
125. Romac JM-J, Ohmuraya M, Bittner C, et al. Transgenic
expression of pancreatic secretory trypsin inhibitor-1
rescues SPINK3-decient mice and restores a normal
pancreatic phenotype. Am J Physiol Gastrointest Liver
Physiol 2010;298:G518G524.
126. Sendler M, Mayerle J, Lerch MM. Necrosis, apoptosis,
necroptosis, pyroptosis: it matters how acinar cells die
during pancreatitis. Cell Mol Gastroenterol Hepatol
2016;2:407408.
127. Gukovskaya AS, Gukovsky I. Which way to die: the
regulation of acinar cell death in pancreatitis by mito-
chondria, calcium, and reactive oxygen species.
Gastroenterology 2011;140:18761880.
128. Gukovskaya AS, Gukovsky I, Algül H, et al. Autophagy,
inammation, and immune dysfunction in the patho-
genesis of pancreatitis. Gastroenterology 2017;
153:12121226.
129. Mareninova OA, Hermann K, French SW, et al. Impaired
autophagic ux mediates acinar cell vacuole formation
and trypsinogen activation in rodent models of acute
pancreatitis. J Clin Invest 2009;119:33403355.
130. Mareninova OA, Sendler M, Malla SR, et al. Lysosome
associated membrane proteins maintain pancreatic acinar
cell homeostasis: LAMP-2 decient mice develop pancre-
atitis. Cell Mol Gastroenterol Hepatol 2015;1:678694.
131. Iwahashi K, Hikita H, Makino Y, et al. Autophagy
impairment in pancreatic acinar cells causes zymogen
granule accumulation and pancreatitis. Biochem Bio-
phys Res Commun 2018;503:25762582.
132. Hashimoto D, Ohmuraya M, Hirota M, et al. Involvement
of autophagy in trypsinogen activation within the
pancreatic acinar cells. J Cell Biol 2008;181:10651072.
133. Gukovsky I, Gukovskaya AS. Impaired autophagy trig-
gers chronic pancreatitis: lessons from pancreas-
specic atg5 knockout mice. Gastroenterology 2015;
148:501505.
134. He S, Huang S, Shen Z. Biomarkers for the detection of
necroptosis. Cell Mol Life Sci 2016;73:21772181.
135. He S, Wang L, Miao L, et al. Receptor interacting protein
kinase-3 determines cellular necrotic response to TNF-
alpha. Cell 2009;137:11001111.
136. Louhimo JM, Steer ML, Perides G. Necroptosis is an
important severity determinant and potential therapeutic
target in experimental severe pancreatitis. Cell Mol
Gastroenterol Hepatol 2016;2:519535.
137. Malik A, Kanneganti T-D. Inammasome activation and
assembly at a glance. J Cell Sci 2017;130:39553963.
1966 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
138. HoqueR,SohailM,MalikA,etal.TLR9andthe
NLRP3 inammasome link acinar cell death with
inammation in acute pancreatitis. Gastroenterology
2011;141:358369.
139. Hoque R, Farooq A, Ghani A, et al. Lactate reduces liver
and pancreatic injury in Toll-like receptor- and
inammasome-mediated inammation via GPR81-
mediated suppression of innate immunity. Gastroenter-
ology 2014;146:17631774.
140. Hoque R, Mehal WZ. Inammasomes in pancreatic
physiology and disease. Am J Physiol Gastrointest Liver
Physiol 2015;308:G643G651.
141. Vince JE, Silke J. The intersection of cell death and
inammasome activation. Cell Mol Life Sci 2016;
73:23492367.
142. Iqbal U, Anwar H, Scribani M. Ringers lactate versus
normal saline in acute pancreatitis: a systematic review
and meta-analysis. J Dig Dis 2018;19:335341.
143. Louhimo J, Steer ML, Perides G. Necroptosis is an
important severity determinant and potential therapeutic
target in experimental severe pancreatitis. Cell Mol
Gastroenterol Hepatol 2016;2:519535.
144. Sendler M, Maertin S, John D, et al. Cathepsin B activity
initiates apoptosis via digestive protease activation in
pancreatic acinar cells and experimental pancreatitis.
J Biol Chem 2016;291:1471714731.
145. Neoptolemos JP, Kemppainen EA, Mayer JM, et al. Early
prediction of severity in acute pancreatitis by urinary
trypsinogen activation peptide: a multicentre study.
Lancet 2000;355:19551960.
146. Selig L, Sack U, Gaiser S, et al. Characterisation of a
transgenic mouse expressing R122H human cationic
trypsinogen. BMC Gastroenterol 2006;6:30.
147. Athwal T, Huang W, Mukherjee R, et al. Expression of
human cationic trypsinogen (PRSS1) in murine acinar
cells promotes pancreatitis and apoptotic cell death. Cell
Death Dis 2014;5:e1165.
148. Gaiser S, Daniluk J, Liu Y, et al. Intracellular activation of
trypsinogen in transgenic mice induces acute but not
chronic pancreatitis. Gut 2011;60:13791388.
149. Archer H, Jura N, Keller J, et al. A mouse model of he-
reditary pancreatitis generated by transgenic expression
of R122H trypsinogen. Gastroenterology 2006;
131:18441855.
150. Dawra R, Sah RP, Dudeja V, et al. Intra-acinar trypsin-
ogen activation mediates early stages of pancreatic
injury but not inammation in mice with acute pancrea-
titis. Gastroenterology 2011;141:22102217.e2.
151. Sah RP, Dudeja V, Dawra RK, et al. Cerulein-induced
chronic pancreatitis does not require intra-acinar acti-
vation of trypsinogen in mice. Gastroenterology 2013;
144:10761085.e2.
152. Ji B, Gaiser S, Chen X, et al. Intracellular trypsin induces
pancreatic acinar cell death but not NF-kappaB activa-
tion. J Biol Chem 2009;284:1748817498.
153. Gukovsky I, Gukovskaya AS, Blinman TA, et al. Early NF-
kappaB activation is associated with hormone-induced
pancreatitis. Am J Physiol Gastrointest Liver Physiol
1998;275:G1402G1414.
154. Steinle AU, Weidenbach H, Wagner M, et al. NF-kappaB/
Rel activation in cerulein pancreatitis. Gastroenterology
1999;116:420430.
155. Baldwin AS. The NF-kappa B and I kappa B proteins:
new discoveries and insights. Annu Rev Immunol 1996;
14:649683.
156. Han B, Logsdon CD. CCK stimulates mob-1 expression
and NF-kappaB activation via protein kinase C and
intracellular Ca(2þ). Am J Physiol Cell Physiol 2000;
278:C344C351.
157. Neuhöfer P, Liang S, Einwächter H, et al. Deletion of IkBa
activates RelA to reduce acute pancreatitis in mice
through up-regulation of Spi2A. Gastroenterology 2013;
144:192201.
158. Rakonczay Z, Hegyi P, Takács T, et al. The role of NF-
kappaB activation in the pathogenesis of acute
pancreatitis. Gut 2008;57:259267.
159. Algül H, Treiber M, Lesina M, et al. Pancreas-specic
RelA/p65 truncation increases susceptibility of acini to
inammation-associated cell death following cerulein
pancreatitis. J Clin Invest 2007;117:14901501.
160. Koike Y, Kanai T, Saeki K, et al. MyD88-dependent
interleukin-10 production from regulatory CD11b
þ
Gr-
1(high) cells suppresses development of acute cerulein
pancreatitis in mice. Immunol Lett 2012;148:172177.
161. Baumann B, Wagner M, Aleksic T, et al. Constitutive
IKK2 activation in acinar cells is sufcient to induce
pancreatitis in vivo. J Clin Invest 2007;117:15021513.
162. Aleksic T, Baumann B, Wagner M, et al. Cellular immune
reaction in the pancreas is induced by constitutively
active IkappaB kinase-2. Gut 2007;56:227236.
163. Li N, Wu X, Holzer RG, et al. Loss of acinar cell IKKa
triggers spontaneous pancreatitis in mice. J Clin Invest
2013;123:22312243.
164. Diakopoulos KN, Lesina M, Wörmann S, et al. Impaired
autophagy induces chronic atrophic pancreatitis in mice
via sex- and nutrition-dependent processes. Gastroen-
terology 2015;148:626638.e17.
165. Cobo I, Martinelli P, Flández M, et al. Transcriptional
regulation by NR5A2 links differentiation and inamma-
tion in the pancreas. Nature 2018;554:533537.
166. Guo L, Sans MD, Hou Y, et al. c-Jun/AP-1 is required for
CCK-induced pancreatic acinar cell dedifferentiation and
DNA synthesis in vitro. Am J Physiol Gastrointest Liver
Physiol 2012;302:G1381G1396.
167. Koh Y-H, Tamizhselvi R, Bhatia M. Extracellular signal-
regulated kinase 1/2 and c-Jun NH2-terminal kinase,
through nuclear factor-kappaB and activator protein-1,
contribute to caerulein-induced expression of sub-
stance P and neurokinin-1 receptors in pancreatic acinar
cells. J Pharmacol Exp Ther 2010;332:940948.
168. Gukovskaya AS, Mouria M, Gukovsky I, et al. Ethanol
metabolism and transcription factor activation in
pancreatic acinar cells in rats. Gastroenterology 2002;
122:106118.
169. Sendler M, Dummer A, Weiss FU, et al. Tumour necrosis
factor asecretion induces protease activation and acinar
cell necrosis in acute experimental pancreatitis in mice.
Gut 2013;62:430439.
May 2019 Genetics and Pathophysiology of Pancreatitis 1967
PANCREATITIS
170. Johnson CD. Persistent organ failure during the rst
week as a marker of fatal outcome in acute pancreatitis.
Gut 2004;53:13401344.
171. Gukovsky I, Li N, Todoric J, et al. Inammation, auto-
phagy, and obesity: common features in the pathogen-
esis of pancreatitis and pancreatic cancer.
Gastroenterology 2013;144:11991209.e4.
172. Habtezion A. Inammation in acute and chronic
pancreatitis. Curr Opin Gastroenterol 2015;31:395399.
173. Liu T, Zhang L, Joo D, et al. NF-kB signaling in inam-
mation. Signal Transduct Target Ther 2017;2:17023.
174. Gukovskaya AS, Gukovsky I, Zaninovic V, et al.
Pancreatic acinar cells produce, release, and respond to
tumor necrosis factor-alpha. Role in regulating cell death
and pancreatitis. J Clin Invest 1997;100:18531862.
175. Sendler M, Weiss F-U, Golchert J, et al. Cathepsin
B-mediated activation of trypsinogen in endocytosing
macrophages increases severity of pancreatitis in mice.
Gastroenterology 2018;154:704718.e10.
176. Piccinini AM, Midwood KS. DAMPening inammation by
modulating TLR signalling. Mediators Inamm 2010;
2010.
177. Zhao Q, Wei Y, Pandol SJ, et al. STING signaling pro-
motes inammation in experimental acute pancreatitis.
Gastroenterology 2018;154:18221835.e2.
178. Gukovskaya AS, Vaquero E, Zaninovic V, et al. Neutro-
phils and NADPH oxidase mediate intrapancreatic
trypsin activation in murine experimental acute pancre-
atitis. Gastroenterology 2002;122:974984.
179. Mayerle J, Schnekenburger J, Krüger B, et al. Extracel-
lular cleavage of E-cadherin by leukocyte elastase during
acute experimental pancreatitis in rats. Gastroenterology
2005;129:12511267.
180. Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil
extracellular traps kill bacteria. Science 2004;303:1532
1535.
181. Kirchner T, Möller S, Klinger M, et al. The impact of
various reactive oxygen species on the formation of
neutrophil extracellular traps. Mediators Inamm 2012;
2012:849136.
182. Leshner M, Wang S, Lewis C, et al. PAD4 mediated
histone hypercitrullination induces heterochromatin
decondensation and chromatin unfolding to form
neutrophil extracellular trap-like structures. Front
Immunol 2012;3:307.
183. Merza M, Hartman H, Rahman M, et al. Neutrophil
extracellular traps induce trypsin activation, inamma-
tion, and tissue damage in mice with severe acute
pancreatitis. Gastroenterology 2015;149:19201931.e8.
184. Schorn C, Janko C, Latzko M, et al. Monosodium urate
crystals induce extracellular DNA traps in neutrophils,
eosinophils, and basophils but not in mononuclear cells.
Front Immunol 2012;3:277.
185. Leppkes M, Maueröder C, Hirth S, et al. Externalized
decondensed neutrophil chromatin occludes pancreatic
ducts and drives pancreatitis. Nat Commun 2016;
7:10973.
186. Schauer C, Janko C, Munoz LE, et al. Aggregated
neutrophil extracellular traps limit inammation by
degrading cytokines and chemokines. Nat Med 2014;
20:511517.
187. Gordon S, Taylor PR. Monocyte and macrophage het-
erogeneity. Nat Rev Immunol 2005;5:953964.
188. Lawrence T, Willoughby DA, Gilroy DW. Anti-inamma-
tory lipid mediators and insights into the resolution of
inammation. Nat Rev Immunol 2002;2:787795.
189. Mareninova OA, Sung K-F, Hong P, et al. Cell death in
pancreatitis: caspases protect from necrotizing pancre-
atitis. J Biol Chem 2006;281:33703381.
190. Perides G, Weiss ER, Michael ES, et al. TNF-alpha-
dependent regulation of acute pancreatitis severity by
Ly-6C(hi) monocytes in mice. J Biol Chem 2011;
286:1332713335.
191. Saeki K, Kanai T, Nakano M, et al. CCL2-induced
migration and SOCS3-mediated activation of macro-
phages are involved in cerulein-induced pancreatitis in
mice. Gastroenterology 2012;142:10101020.e9.
192. Fink SL, Cookson BT. Caspase-1dependent pore for-
mation during pyroptosis leads to osmotic lysis of
infected host macrophages. Cell Microbiol 2006;8:1812
1825.
193. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host
cell death and inammation. Nat Rev Microbiol 2009;
7:99109.
194. Guo H, Callaway JB, Ting JP-Y. Inammasomes:
mechanism of action, role in disease, and therapeutics.
Nat Med 2015;21:677687.
195. Marrache F, Tu SP, Bhagat G, et al. Overexpression of
interleukin-1beta in the murine pancreas results in
chronic pancreatitis. Gastroenterology 2008;135:1277
1287.
196. Xue J, Sharma V, Hsieh MH, et al. Alternatively activated
macrophages promote pancreatic brosis in chronic
pancreatitis. Nat Commun 2015;6:7158.
197. Criscimanna A, Coudriet GM, Gittes GK, et al. Activated
macrophages create lineage-specic microenviron-
ments for pancreatic acinar- and b-cell regeneration in
mice. Gastroenterology 2014;147:11061118.e11.
198. Sendler M, Beyer G, Mahajan UM, et al. Complement
component 5 mediates development of brosis, via
activation of stellate cells, in 2 mouse models of chronic
pancreatitis. Gastroenterology 2015;149:765776.e10.
Received September 7, 2018. Accepted November 16, 2018.
Reprint requests
Address requests for reprints to: Markus M. Lerch, MD, FRCP, Department of
Medicine A, University Medicine Greifswald, Ernst-Moritz-Arndt-University
Greifswald, Ferdinand-Sauerbruch-Strasse 1, 17475 Greifswald, Germany.
e-mail: lerch@uni-greifswald.de.
Acknowledgments
This article is dedicated to the memory of Walter Halangk, PhD, to honor his
work as a pioneer unraveling the pathophysiology of pancreatitis.
Conicts of interest
The authors disclose no conicts.
Funding
This study was funded by the PePPP Center of Excellence (MV ESF/14-BM-
A55-0045/16; ESF MV V-630-S-150-2012/132/133; DFG-CRC 1321.-P14;
and DFG SE 2702/2-1) and the National Institutes of Health (grants R01
DK058088 and R01 DK117809 to Miklós Sahin-Tóth).
1968 Mayerle et al Gastroenterology Vol. 156, No. 7
PANCREATITIS
Supplementary Table 1.Genetic Risk Factors in Chronic Pancreatitis
Gene/protein Mutation Functional effect Phenotype
PRSS1/cationic
trypsinogen
p.R122C, p.R122H Increased trypsinogen activation owing to
prevention of CTRC-mediated
degradation
Autosomal dominant HP,
familial pancreatitis, or
sporadic CP
pA16V, p.P17T, p.N29I Increased CTRC-dependent trypsinogen
autoactivation
p.D19A, p.D21A, p.D22G,
p.K23R, p.K23_I24insIDK
Increased CTRC-independent trypsinogen
autoactivation
c.204C>A Decreased trypsinogen expression Protection against alcoholic
CP
p.C139F, p.C139S,p.L104P,
p.R116C, p.G208A
Decreased secretion, intracellular retention,
and increased ER stress owing to mis-
folding
Sporadic or familial CP
PRSS2/anionic
trypsinogen
p.G191R Introduction of new trypsin cleavage site,
increased autocatalytic inactivation
Protection against CP
SPINK1/trypsin inhibitor p.N34S unknown Increased susceptibility for CP
c.194þ2T>C Skipping of exon 3 resulting in lower SPINK1
levels
CTRC/chymotrypsin C p.A73T Secretion defect Increased susceptibility for CP
p.K247_R254del Inactive CTRC
p.R254W CTRC degradation by trypsin
p.V235I Decreased CTRC activity
p.G60¼Decreased CTRC mRNA
CTRB12/chymotrypsin B1
and B2
Inversion at CTRB1CTRB2
locus
Changes expression ratio of CTRB1 and
CTRB2
Protection against CP
CPA1/carboxypeptidase
A1
p.S282P, p.N256K Proenzyme mis-folding, secretion defect,
intracellular retention, and ER stress
Sporadic and familial CP
CEL/carboxyl ester lipase Single-nucleotide deletions Protein retention and ER stress Exocrine pancreatic
insufciency associated
with MODY8
CEL-HYB1 Protein retention and ER stress? Increased susceptibility for CP
CFTR/cystic brosis
transmembrane
conductance regulator
p.F508del, p.R117H Loss of CFTR function due to mis-folding
and degradation or disrupted channel
activity
Increased susceptibility for CP
CLDN2/claudin 2 CLDN2MORC4 variants Tight junction defect? Increased susceptibility for CP
CASR/calcium-sensing
receptor
p.R990G Altered bicarbonate and uid secretion from
disrupted ductal calcium sensing?
Increased susceptibility for
CP?
p.A986S
May 2019 Genetics and Pathophysiology of Pancreatitis 1968.e1
... Similar to CFTR mutations, the exact contribution of CaSR variants to pancreatitis risk remains controversial, although it has been demonstrated that these variants may impair calcium sensing and promote higher ductal Ca 2+ levels [22]. Recent studies in the literature reported that common CaSR variants do not alter the risk of CP and should not be considered as responsible for genetic risk in subjects suffering from pancreatitis [23,24]. ...
Article
Full-text available
Chronic pancreatitis is often secondary to alcohol abuse, but pancreatitis with no other aetiology is frequently associated with variants in genes encoding proteins related to zymogen granule activation. Our goal was to identify genomic variants in a patient by analyzing an extended panel of genes associated with the intra-pancreatic activation of the trypsin pathway. A 23-year-old woman was addressed at our institution because of chronic pancreatitis of unknown aetiology presenting recurrent episodes since she was the age of four. Next Generation Sequencing was performed to analyze a panel of nine genes associated with pancreatitis (CaSR, CFTR, CPA1, CTRC, CTSB, KRT8, PRSS1, PRSS2, and SPINK1). Three missense variants were found: p.Leu997Phe, maternally inherited, in the CFTR gene; p.Ile73Phe, paternally inherited, in the SPINK1 gene; and p.Phe790Ser, a de novo variant, in the CaSR gene. They were classified, respectively as probably benign, a Variant of Uncertain Significance, and the last one, which has never been described in the literature, as likely being pathogenic following American College of Medical Genetics and Genomics standard guidelines. Extensive intra-pancreatic activation of trypsin pathway gene sequencing detected rare variants that were not found with other gene screening and showed that variants in different genes may interact in contributing to the onset of the pancreatitis phenotype.
... Furthermore, recurrence occurs in 21% of patients after the first episode of AP, and chronic pancreatitis (CP) develops in 36% of patients after recurrent AP (RAP) [10]. Over the past 2 decades, clinical data accumulated from human studies have confirmed that AP, RAP, and CP are considered to be a disease continuum [11]. However, it is also important to recognize that patients with AP may not necessarily eventually develop CP, and similarly patients with CP may have subclinical AP episodes or no history of AP at all [12]. ...
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.
... Острый билиарный панкреатит (ОБП) является осложнением желчнокаменной болезни и возникает вследствие нарушения оттока желчи и панкреатического сока, относится к группе мультифакторных заболеваний, развитие которых наступает вследствие взаимодействия генетических и средовых факторов. Поскольку в патогенезе заболевания ведущую роль выполняют ферменты поджелудочной железы, калликреин-кининовая система и цитокины, то основное количество исследований в мире посвящено изуче-нию роли генов, ответственных за синтез именно этих ферментов [1][2][3]. К средовым факторам риска относят употребление жирной, жареной, острой пищи, стимулирующей экзокринную функцию поджелудочной железы, недостаточное употребление свежих овощей и фруктов, богатых витаминами и антиоксидантами, а также злоупотребление алкоголем и курение [4,5]. ...
Article
Aim. To investigate the role of single nucleotide polymorphisms (SNPs) rs13041792, rs1801310, and rs6088660 in the GSS gene and environmental factors in the development of acute biliary pancreatitis (ABP) and its complications. Materials and methods. The material for the study was blood samples obtained from 84 patients with ABP and 573 healthy individuals. Both groups were comparable in terms of gender and age. To diagnose ABP, we used the clinical guidelines recommended by the working group of the Russian Society of Surgeons. DNA was isolated by phenol / chloroform extraction. Multiplex genotyping of SNPs was performed by the iPLEX assay on the MALDITOF MassARRAY-4 genetic analyzer. Statistical data processing was performed using Statistica 10 and SNPStats software. Results. We found that insufficient consumption of fresh vegetables and fruits increased the probability of ABP in carriers of genotypes G/A-A/A at rs1801310 in GSS ( р = 0.02). The analysis revealed the association of the T allele at rs6088660 with the odds for developing acute pancreatitis ( р = 0.007) and digestive fistulas ( р = 0.02). A high probability of death was associated with rs1801310 (G/A genotype, р = 0.002) and rs6088660 (C/T genotype, р = 0.01) in the GSS gene. Conclusion. SNPs rs6088660 and rs1801310 in the GSS gene can be used to predict the course of ABP.
... Pancreatic cancer is one of the most common malignant gastrointestinal tumors worldwide, and its incidence and mortality rates are increasing each year [1]. The current mainstay of pancreatic cancer treatment is surgical resection, and early surgery has been shown to increase the likelihood of successful resection, with postoperative 5-year survival rates ranging from 70% to 85% [2]. ...
Article
BACKGROUND Advanced pancreatic cancer is resistant to chemotherapeutic drugs, resulting in limited treatment efficacy and poor prognosis. Combined administration of the chemotherapeutic gemcitabine and erlotinib is considered a potential first-line treatment for advanced pancreatic cancer. However, their comparative benefits and potential risks remain unclear. AIM To assess the clinical efficacy and safety of erlotinib combined with other chemotherapy regimens for the treatment of advanced pancreatic cancer. METHODS Literature on the clinical efficacy and safety of erlotinib combined with chemotherapy for advanced pancreatic cancer was retrieved through an online search. The retrieved literature was subjected to a methodological qualitative assessment and was analyzed using the RevMan 5.3 software. Ten randomized controlled trials involving 2444 patients with advanced pancreatic cancer were included in the meta-analysis. RESULTS Compared with chemotherapeutic treatment, erlotinib combined with chemotherapy significantly prolonged the progression-free survival time of pancreatic cancer patients [hazard ratio (HR) = 0.78, 95%CI: 0.66–0.92, P = 0.003]. Meanwhile, the overall survival (HR= 0.99, 95%CI: 0.72–1.37, and P = 0.95) and disease control rate (OR = 0.93, 95%CI: 0.45–0.91, P = 0.84) were not significantly favorable. In terms of safety, the erlotinib and chemotherapy combination was associated with a significantly higher risk of diarrhea (OR = 3.59, 95%CI: 1.63–7.90, P < 0.05) and rash (OR = 3.63, 95%CI: 1.64–8.01, P < 0.05) compared with single-agent chemotherapy. Moreover, the risk of vomiting (OR = 1.27, 95%CI: 0.62–2.59, P = 0.51), regurgitation/anorexia (OR = 1.61, 95%CI: 0.25–10.31, P = 0.62), and infection (OR = 0.72, 95%CI: 0.28-1.87, P = 0.50) were not significant in either group. CONCLUSION Compared with a single chemotherapeutic modality, erlotinib combined with gemcitabine can prolong progression-free survival in pancreatic cancer, but does not improve survival benefit or disease control rate, and can increase the risk of diarrhea and rash.
... thereby exacerbating CP [5]. An initial event in CP is cell death resulting from the abnormal activation of trypsinogen in acinar cells [6]. Similarly, the activation of digestive enzymes in the pancreas is the underlying cause of acute pancreatitis (AP) [7]. ...
Article
Full-text available
Acute pancreatitis (AP) and chronic pancreatitis (CP) are considered to be two separate pancreatic diseases in most studies, but some clinical retrospective analyses in recent years have found some degree of correlation between the two in actual treatment, however, the exact association is not clear. In this study, bioinformatics analysis was utilized to examine microarray sequencing data in mice, with the aim of elucidating the critical signaling pathways and genes involved in the progression from AP to CP. Differential gene expression analyses on murine transcriptomes were conducted using the R programming language and the R/Bioconductor package. Additionally, gene network analysis was performed using the STRING database to predict correlations among genes in the context of pancreatic diseases. Functional enrichment and gene ontology pathways common to both diseases were identified using Metascape. The hub genes were screened in the cytoscape algorithm, and the mRNA levels of the hub genes were verified in mice pancreatic tissues of AP and CP. Then the drugs corresponding to the hub genes were obtained in the drug-gene relationship. A set of hub genes, including Jun, Cd44, Epcam, Spp1, Anxa2, Hsp90aa1, and Cd9, were identified through analysis, demonstrating their pivotal roles in the progression from AP to CP. Notably, these genes were found to be enriched in the Helper T-cell factor (Th17) signaling pathway. Up-regulation of these genes in both AP and CP mouse models was validated through quantitative real-time polymerase chain reaction (qRT-PCR) results. The significance of the Th17 signaling pathway in the transition from AP to CP was underscored by our findings. Specifically, the essential genes driving this progression were identified as Jun, Cd44, Epcam, Spp1, Anxa2, Hsp90aa1, and Cd9. Crucial insights into the molecular mechanisms underlying pancreatitis progression were provided by this research, offering promising avenues for the development of targeted therapeutic interventions.
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
Regulated exocytosis is initiated by increased Ca²⁺ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca²⁺ directly released from acidic Ca²⁺ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca²⁺ regulator protein integral to the membrane of secretory granules that controlled Ca²⁺ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca²⁺ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca²⁺ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.
Article
Objective Currently, there is no cure for chronic pancreatitis (CP). Germline loss-of-function variants in SPINK1 (encoding trypsin inhibitor) are common in patients with CP and are associated with acute attacks and progression of the disease. This preclinical study was conducted to explore the potential of adeno-associated virus type 8 (AAV8)-mediated overexpression of human SPINK1 ( hSPINK1 ) for pancreatitis therapy in mice. Design A capsid-optimised AAV8-mediated hSPINK1 expression vector (AAV8- hSPINK1 ) to target the pancreas was constructed. Mice were treated with AAV8- hSPINK1 by intraperitoneal injection. Pancreatic transduction efficiency and safety of AAV8- hSPINK1 were dynamically evaluated in infected mice. The effectiveness of AAV8- hSPINK1 on pancreatitis prevention and treatment was studied in three mouse models (caerulein-induced pancreatitis, pancreatic duct ligation and Spink1 c.194+2T>C mouse models). Results The constructed AAV8- hSPINK1 vector specifically and safely targeted the pancreas, had low organ tropism for the heart, lungs, spleen, liver and kidneys and had a high transduction efficiency (the optimal expression dose was 2×10 ¹¹ vg/animal). The expression and efficacy of hSPINK1 peaked at 4 weeks after injection and remained at significant level for up to at least 8 weeks. In all three mouse models, a single dose of AAV8 -hSPINK1 before disease onset significantly alleviated the severity of pancreatitis, reduced the progression of fibrosis, decreased the levels of apoptosis and autophagy in the pancreas and accelerated the pancreatitis recovery process. Conclusion One-time injection of AAV8 -hSPINK1 safely targets the pancreas with high transduction efficiency and effectively ameliorates pancreatitis phenotypes in mice. This approach is promising for the prevention and treatment of CP.
Article
Full-text available
Background & aims: Estimates of disease burden can inform national health priorities for research, clinical care, and policy. We aimed to estimate health care use and spending among gastrointestinal (GI) (including luminal, liver, and pancreatic) diseases in the United States. Methods: We estimated health care use and spending based on the most currently available administrative claims from commercial and Medicare Supplemental plans, data from the GI Quality Improvement Consortium Registry, and national databases. Results: In 2015, annual health care expenditures for gastrointestinal diseases totaled $135.9 billion. Hepatitis ($23.3 billion), esophageal disorders ($18.1 billion), biliary tract disease ($10.3 billion), abdominal pain ($10.2 billion), and inflammatory bowel disease ($7.2 billion) were the most expensive. Yearly, there were more than 54.4 million ambulatory visits with a primary diagnosis for a GI disease, 3.0 million hospital admissions, and 540,500 all-cause 30-day readmissions. There were 266,600 new cases of GI cancers diagnosed and 144,300 cancer deaths. Each year, there were 97,700 deaths from non-malignant GI diseases. An estimated 11.0 million colonoscopies, 6.1 million upper endoscopies, 313,000 flexible sigmoidoscopies, 178,400 upper endoscopic ultrasound examinations, and 169,500 endoscopic retrograde cholangiopancreatography procedures were performed annually. Among average-risk persons aged 50-75 years who underwent colonoscopy, 34.6% had 1 or more adenomatous polyps, 4.7% had 1 or more advanced adenomatous polyps, and 5.7% had 1 or more serrated polyps removed. Conclusions: GI diseases contribute substantially to health care use in the United States. Total expenditures for GI diseases are $135.9 billion annually-greater than for other common diseases. Expenditures are likely to continue increasing.
Article
Full-text available
Key points: Giant trypsin-containing endocytic vacuoles are formed in pancreatic acinar cells stimulated with inducers of acute pancreatitis. F-actin envelops endocytic vacuoles and regulates their properties. Endocytic vacuoles can rupture and release their content into the cytosol of the acinar cells. Endocytic vacuoles can fuse with the plasma membrane of acinar cells and exocytose their content. Abstract: Intrapancreatic activation of trypsinogen is an early event in and hallmark of the development of acute pancreatitis. Endocytic vacuoles, which form by disconnection and transport of large post-exocytic structures, are the only resolvable sites of the trypsin activity in live pancreatic acinar cells. In this study we characterised dynamics of endocytic vacuole formation induced by physiological and pathophysiological stimuli and visualised a prominent actin coat that completely or partially surrounded endocytic vacuoles. An inducer of acute pancreatitis taurolithocholic acid 3-sulfate (TLC-S) and supramaximal concentrations of cholecystokinin (CCK) triggered formation of giant (more than 2.5 μM in diameter) endocytic vacuoles. We discovered and characterised intracellular rupture of endocytic vacuoles and fusion of endocytic vacuoles with basal and apical regions of the plasma membrane. Experiments with specific protease inhibitors suggest that the rupture of endocytic vacuoles is unlikely to be induced by trypsin or cathepsin B. Perivacuolar filamentous actin (observed on the surface of approximately 30% of endocytic vacuoles) may play a stabilising role by preventing rupture of the vacuoles and fusion of the vacuoles with the plasma membrane. The rupture and fusion of endocytic vacuoles allow trypsin to escape the confinement of a membrane-limited organelle, gain access to intracellular and extracellular targets and initiate autodigestion of the pancreas, a crucial pathophysiological event. This article is protected by copyright. All rights reserved.
Article
Full-text available
Merely touching the pancreas can lead to premature zymogen activation and pancreatitis but the mechanism is not completely understood. Here we demonstrate that pancreatic acinar cells express the mechanoreceptor Piezo1 and application of pressure within the gland produces pancreatitis. To determine if this effect is through Piezo1 activation, we induce pancreatitis by intrapancreatic duct instillation of the Piezo1 agonist Yoda1. Pancreatitis induced by pressure within the gland is prevented by a Piezo1 antagonist. In pancreatic acinar cells, Yoda1 stimulates calcium influx and induces calcium-dependent pancreatic injury. Finally, selective acinar cell-specific genetic deletion of Piezo1 protects mice against pressure-induced pancreatitis. Thus, activation of Piezo1 in pancreatic acinar cells is a mechanism for pancreatitis and may explain why pancreatitis develops following pressure on the gland as in abdominal trauma, pancreatic duct obstruction, pancreatography, or pancreatic surgery. Piezo1 blockade may prevent pancreatitis when manipulation of the gland is anticipated.
Article
Full-text available
Significance Much of the inherited susceptibility to pancreatic cancer remains unexplained. Germline variants that cause protein misfolding and impaired secretion of pancreatic enzymes such as CPA1 (encoding carboxypeptidase A1) can cause pancreatic acinar cell endoplasmic reticulum (ER) stress. We investigated the hypothesis that pancreatic cancer could arise from germline variants in genes encoding pancreatic secretory enzymes that induce pancreatic acinar cell stress. We find ∼1% of 1,579 patients with pancreatic cancer vs. 1 of 2,012 controls have germline variants in the genes encoding CPA1 and CPB1 (carboxypeptidase B1) that impair secretion of its protein product and induce ER stress. These findings implicate pancreatic acinar cell stress as a mechanism of pancreatic cancer susceptibility.
Article
Full-text available
Chronic inflammation increases the risk of developing one of several types of cancer. Inflammatory responses are currently thought to be controlled by mechanisms that rely on transcriptional networks that are distinct from those involved in cell differentiation. The orphan nuclear receptor NR5A2 participates in a wide variety of processes, including cholesterol and glucose metabolism in the liver, resolution of endoplasmic reticulum stress, intestinal glucocorticoid production, pancreatic development and acinar differentiation. In genome-wide association studies, single nucleotide polymorphisms in the vicinity of NR5A2 have previously been associated with the risk of pancreatic adenocarcinoma. In mice, Nr5a2 heterozygosity sensitizes the pancreas to damage, impairs regeneration and cooperates with mutant Kras in tumour progression. Here, using a global transcriptomic analysis, we describe an epithelial-cell-autonomous basal pre-inflammatory state in the pancreas of Nr5a2+/-mice that is reminiscent of the early stages of pancreatitis-induced inflammation and is conserved in histologically normal human pancreases with reduced expression of NR5A2 mRNA. In Nr5a2+/-mice, NR5A2 undergoes a marked transcriptional switch, relocating from differentiation-specific to inflammatory genes and thereby promoting gene transcription that is dependent on the AP-1 transcription factor. Pancreatic deletion of Jun rescues the pre-inflammatory phenotype, as well as binding of NR5A2 to inflammatory gene promoters and the defective regenerative response to damage. These findings support the notion that, in the pancreas, the transcriptional networks involved in differentiation-specific functions also suppress inflammatory programmes. Under conditions of genetic or environmental constraint, these networks can be subverted to foster inflammation.
Article
Intra-pancreatic activation of the digestive proteases trypsin and chymotrypsin is an early event in the development of pancreatitis. Human genetic studies indicate that chymotrypsin controls trypsin activity via degradation, but there is no evidence of this from animal models. We used CRISPR-Cas9 to disrupt the chymotrypsinogen B1 gene (Ctrb1) in C57BL/6N mice and induced pancreatitis in CTRB1-deficient and C57BL/6N (control) mice by administration of cerulein. CTRB1-deficient mice given cerulein had significant increases in intra-pancreatic trypsin activity and developed more severe pancreatitis compared with control mice. CTRB1 therefore protects against secretagogue-induced pancreatitis by reducing trypsin activity. Protease inhibitors developed for treatment of pancreatitis should be designed to target trypsin but not chymotrypsin.
Article
Objective Chronic pancreatitis is a progressive, relapsing inflammatory disorder of the pancreas, which often develops in the background of genetic susceptibility. Recently, loss-of-function mutations in CPA1, which encodes the digestive enzyme carboxypeptidase A1, were described in sporadic early onset cases and in hereditary pancreatitis. Mutationinduced misfolding of CPA1 and associated endoplasmic reticulum (ER) stress was suggested as potential disease mechanism; however, in vivo evidence has been lacking. The objective of the present study was to create a mouse model that recapitulates features of CPA1-associated chronic pancreatitis. Design We knocked-in the most frequently occurring p.N256K human CPA1 mutation to the mouse Cpa1 locus. Mutant mice were with respect to pancreas pathology and ER stress and C57BL/ and CPA1 null control mice. Results In the CPA1 N256K mutant mice, we observed hallmarks of chronic pancreatitis that included progressive acinar cell atrophy, inflammatory cell infiltration, fibrosis and acinar-ductal metaplasia. In contrast, similarly to the C57BL/6N mice, the CPA1 null control strain exhibited no signs of pancreatic disease. Mutation p.N256K induced misfolding of mouse CPA1 and resulted in elevated expression of ER stress markers Hspa5 (BiP) and Ddit3 (CHOP) both in cell culture and mutant mice. Conclusion The results offer categorical evidence that CPA1 mutations elicit enzyme misfolding and cause chronic pancreatitis via an ER stress-related mechanism.
Article
Basal autophagy degrades many kinds of proteins and organelles to maintain quality and plays important roles in cellular homeostasis. However, the impact of basal autophagy on zymogen granules in pancreatic acinar cells is unknown. In the present study, we examined the influence of autophagy impairment in acinar cells on zymogen granules and homeostasis of the pancreas, using mice with pancreas-specific autophagy impairment (Pdx1-Cre+/-Atg7fl/fl mice). The number of zymogen granules in acinar cells from these mice did not differ from that in acinar cells from their wild-type littermates at 3 weeks of age. However, the number of zymogen granules in acinar cells drastically increased at 4 weeks of age in mice with pancreas-specific autophagy impairment. In addition to the increased number of zymogen granules, serum lipase was elevated, and the pancreas became oedematous at 4 weeks of age, suggesting pancreatitis. After 5 weeks of age, acinar cell death was accelerated, and several histological features of chronic pancreatitis were observed, including glandular atrophy and pseudotubular complexes with fibrotic changes. In conclusion, the impairment of pancreas-specific basal autophagy caused spontaneous zymogen granule accumulation in acinar cells and pancreatitis, which eventually led to chronic pancreatitis.
Article
Introduction Acute Pancreatitis (AP) is one of the most common causes of hospitalization in the United States. Aggressive intravenous hydration with crystalloids is the first step in management, and is associated with improved survival. Guidelines are unclear regarding the choice of crystalloids. Normal saline (NS) is most commonly used, but recent studies have shown Ringer's lactate (RL) to be associated with better outcomes. Methods A comprehensive literature review was conducted by searching the Embase, MEDLINE, Pubmed, and Google Scholar databases through December 2017 to identify all studies that compared the use of NS with RL for the management of acute pancreatitis. Two independent reviewers extracted data and assessed the quality of publications; a third investigator resolved any discrepancies. Results Five studies, three randomized controlled trials and two retrospective cohort studies, including 428 patients, were included in this analysis. Mortality trended lower in the RL group, but was not statistically significant (pooled odds ratio 0.61 (0.28‐1.29; P=0.20)). Patients in the RL group had significantly decreased odds of developing systemic inflammatory response (SIRS) at 24 hours (pooled odds ratio 0.38 (0.15‐0.98; P=0.05)). Conclusion This study demonstrated that RL has anti‐inflammatory effects and is associated with decreased odds of persistent SIRS at 24 hours, which is a marker of severe disease in patients with AP. Although mortality trended lower in RL group, it did not achieve statistical significance and hence larger randomized controlled trials are needed to evaluate this association.