You are on page 1of 17

SCIENCE ADVANCES | RESEARCH ARTICLE

IMMUNOLOGY Copyright © 2021


The Authors, some
Adipose saturation reduces lipotoxic systemic rights reserved;
exclusive licensee
inflammation and explains the obesity paradox American Association
for the Advancement
of Science. No claim to
Biswajit Khatua1, Bara El-Kurdi1, Krutika Patel1, Christopher Rood2, Pawan Noel1, original U.S. Government
Michael Crowell1, Jordan R. Yaron1, Sergiy Kostenko1, Andre Guerra1, Douglas O. Faigel1, Works. Distributed
Mark Lowe3, Vijay P. Singh1* under a Creative
Commons Attribution
Obesity sometimes seems protective in disease. This obesity paradox is predominantly described in reports from NonCommercial
the Western Hemisphere during acute illnesses. Since adipose triglyceride composition corresponds to long-term License 4.0 (CC BY-NC).
dietary patterns, we performed a meta-analysis modeling the effect of obesity on severity of acute pancreatitis, in
the context of dietary patterns of the countries from which the studies originated. Increased severity was noted in
leaner populations with a higher proportion of unsaturated fat intake. In mice, greater hydrolysis of unsaturated
visceral triglyceride caused worse organ failure during pancreatitis, even when the mice were leaner than those
having saturated triglyceride. Saturation interfered with triglyceride’s interaction and lipolysis by pancreatic
triglyceride lipase, which mediates organ failure. Unsaturation increased fatty acid monomers in vivo and aqueous

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


media, resulting in greater lipotoxic cellular responses and organ failure. Therefore, visceral triglyceride saturation
reduces the ensuing lipotoxicity despite higher adiposity, thus explaining the obesity paradox.

INTRODUCTION We therefore approached the obesity paradox by examining vis-


While quantitative parameters such as body mass index (BMI) (1), waist ceral fat composition as influenced by dietary fat composition (29).
circumference, and amount of visceral adipose (2, 3) are well-studied Epidemiologically, all clinical reports up to 2017 associating AP severity
risk factors for acute disease severity, the impact of adipose compo- with BMI were grouped by cutoff BMI, and the dietary fat composi-
sition on severity is unclear. Organ failure, the hallmark of acute tion representative of the country from which the study originated
pancreatitis (AP) severity, has been associated with long-chain non- was studied. The dietary fat data were based on publicly available
esterified fatty acid (NEFA) lipotoxicity in humans (4–7). Clues to information archived by the Food and Agriculture Organization
the role of adipose composition in severity come from studies from (FAO) and were classified as saturated or unsaturated based on the
Western populations showing a BMI of >30 mostly, but not always presence of double bonds in long-chain (>12 carbon) NEFA com-
(2, 3) associated with severity, while in Eastern populations, much posing the dietary triglycerides (described in Methods). After analyz-
lower BMIs, sometimes ≥23 (8, 9), are associated with severity. This ing the human metadata, we tested the plausibility of fat composition
“obesity paradox” (10) also occurs in other acute scenarios such as burns affecting the outcomes of pancreatitis by altering mouse visceral fat
(11), acute heart failure (12), after trauma (13), cardiovascular surgery composition to replicate human visceral fat (25, 26, 29). Unexpectedly,
(14), and during critical illnesses (15) in which elevated pancreatic the human and experimental models cumulatively showed that the
enzymes (16–18) and long-chain NEFA (19, 20) have been associated U.S. Food and Drug Administration (FDA)–recommended higher
with severity. More recent studies associate such an enzyme leak (21) unsaturated fat intake (https://health.gov/dietaryguidelines/2015/
and dietary fat composition (22), along with elevated NEFA levels, resources/2015-2020_Dietary_Guidelines.pdf; https://www.fda.gov/
to the severity (23) and mortality from coronavirus disease 2019 files/food/published/Food-Labeling-Guide-%28PDF%29.pdf, appen-
(COVID-19) (22, 24). However, the mechanisms determining NEFA dix F) increased the risk of severe AP (SAP). We therefore investigated
generation and how NEFA mediate outcomes such as the cytokine the mechanistic basis of this finding and noted that saturation of a
storm (24) and organ failure (19, 20, 22) are unclear. triglyceride makes its interaction with PNLIP energetically unfavor-
Most individual studies supporting the obesity paradox come able and reduces its lipolysis. Moreover, in general, unsaturated NEFAs
from the Western Hemisphere (12–14), where dietary and adipose achieved higher monomeric concentrations in vivo and aqueous
fat are more saturated (25, 26). We therefore aimed at understanding systems, consequently eliciting stronger biological responses and
this problem in different populations, by modeling AP, wherein the causing worse injury and organ failure. These features may explain
pancreatic lipases leak into the surrounding fat (6, 7, 27) and hydrolyze how obese populations with higher visceral fat saturation may
the neutral lipids (7). Principal among these lipases is pancreatic sometimes be protected from severe disease.
triglyceride lipase (PNLIP), which enters visceral adipocytes by various
mechanisms (7) and mediates lipotoxic systemic injury along with
organ failure (7, 28) via the generated long-chain NEFA (7, 28) inhib- RESULTS
iting mitochondrial complexes I and V (6). However, the impact of Higher saturated fat intake is associated with SAP at
long-chain NEFA saturation on systemic inflammation is unknown. a higher BMI
As seen in Fig. 1A and fig. S1, 20 reports from 11 countries used a
1
BMI cutoff of ≥30 to stratify SAP. Six of these (blue countries and
Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA. 2Saint Louis University text in table) reported no increased risk (2, 3, 30–34), while the 14
School of Medicine, Saint Louis, MO, USA. 3Department of Pediatrics, Washington
University School of Medicine, Saint Louis, MO, USA. (golden text and green countries) reported an increased risk of SAP
*Corresponding author. Email: singh.vijay@mayo.edu at BMI of ≥30 (1, 6, 35–44). Six reports used a cut off BMI of 25 or

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 1 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


Fig. 1. Relationship of human SAP to cutoff BMI and composition of dietary fat intake. (A) Studies using a BMI cutoff of ≤25 are shown in pink, those associating SAP
with a BMI of ≥30 are shown in gold, and studies showing a BMI of >30 was not associated with severe AP (SAP) are show shown in blue. The respective countries from
which the study originated are shown in pink, green, and blue, with a checkered pattern for countries with studies showing different outcomes. Box plot comparing the
per capita per year saturated fat (from dairy, cattle, and palm oil) consumption in countries with BMI cutoffs of >30 and those with ≤25 BMI (B) and %UFA in dietary fat
intake (C). (D) Meta-analysis showing OR for SAP for studies using a BMI cutoff of ≤25 (pink background) and those using a BMI cutoff of >30 (blue-yellow background).
The overall OR is shown at the bottom (orange background).

less (8, 9, 45–49). Adult AP typically occurs after the third decade was not detected on the basis of a Funnel plot (fig. S1C), an Egger’s
(50, 51). Since the composition of visceral fat necrosed during AP regression, or a Begg and Mazumdar rank correlation. There were
may be influenced by the dietary fat composition over the preced- no differences in age, sex distribution, or etiology of AP between the
ing years (26), we analyzed dietary fat composition of different two groups (fig. S1). While the effect of genes and comorbidities on
countries shown in Fig. 1A. The per capita fat consumption was %SAP cannot be commented on in these data, meta-regression
calculated by averaging the yearly data (1970 to 2011) for each showed that %unsaturated fatty acid (UFA) was able to explain 33%
country. Countries with a BMI cutoff of ≥30 had higher per capita of the heterogeneity in the rate of SAP, and neither age, AP etiology,
saturated fat consumption (Fig. 1B and fig. S1). While the amount nor per capita gross domestic product (GDP) correlated with SAP.
of unsaturated fat consumption was the same (fig. S3A), unsatu- Furthermore, while per capita GDP of countries averaged for 5 years
rated fat comprised a higher percentage of fat intake in the coun- preceding publication was significantly lower in countries reporting
tries with reports having a cutoff BMI of ≤25 (pink countries; SAP at BMIs of ≤25 ($15,768 ± 14,624 versus $32,272 ± 17,007,
Fig. 1, A and C). Overall, there was a moderate correlation between P = 0.018), neither mortality (5.2 ± 6.9 versus 6.7 ± 4.3%, P = 0.698)
the percentage of patients with SAP and the percentage of unsatu- nor the proportion of patients with SAP (24 ± 12.5 versus 20.7 ±
rated fat intake (fig. S3B). On meta-­analysis (Fig. 1D), a significantly 8.4%, P = 0.892) was different in the two BMI cutoff groups. Therefore,
increased risk of severity was noted for cutoff BMIs of ≤25 [pink despite differences in income, the proportion of SAP and mortality
shade, odds ratio (OR) 2.8, CI 1.3 to P = 0.008] and also BMI of >30 were unchanged. This implied that quality of care was not differ-
(blue and greens, OR 2.7, CI 1.8 to 3.8, P < 0.001). Publication bias ent in the two groups of countries. Overall, these studies supported

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 2 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

that severe pancreatitis occurred at a lower BMI in countries with a concentrations in common FDA-recommended dietary fats (52)
lower dietary saturated fatty acid (SFA) intake. Since we are not aware (e.g., cooking oils) and comprises a high proportion in pancreatic
of the literature on how diet would affect the genetic background of fat necrosis (FN) (5, 6). Being essential, LA’s concentrations in
a population over several generations to alter the severity of pan- visceral fat parallel dietary intake (29). We thus formulated a diet
creatitis, we therefore went on to experimentally study whether and enriched in LA (70% LA in a 45% fat diet) to represent high %UFA
how altering visceral fat composition affected the development of intake in dietary fat (red box in fig. S4A and Fig. 2A1, red columns)
SAP in the commonly used caerulein (CER) model of AP, which, in (26, 52). Similarly, a diet enriched in saturated fat was formulated,
lean C57BL6 normal chow-fed mice, remains mild, self-limited, and which had palmitic acid (PA; C16:0, PA 68% of a 47% fat diet; fig.
without organ failure (27). S4B, green rectangle) replicating the 65 to 70% saturation of dairy
fat. These diets spanned the range of %UFA intake in humans (16% in
Dietary unsaturated fat results in unsaturated visceral fat Australia and 79% in Japan), as shown in fig. S3A.
and worsens AP more than higher amounts of saturated Mice fed these diets had a similar food intake (6.5 ± 1.4 g/day of
visceral fat UFA diet or 6.3 ± 0.5 g/day of SFA diet). This altered their visceral
We first aimed at recapitulating the human studies showing that triglyceride composition, with LA and PA increasing to 40 to 45%
dietary fat composition can change adipose tissue composition in (Fig. 2A1 and detailed in fig. S5) in the UFA- and SFA-fed groups,
an animal model (26). Linoleic acid (LA; C18:2) is an unsaturated respectively. We previously showed that a normal chow diet with
essential fatty acid (i.e., only available through diet) present at high 5% fat diet (Purina 5053), which contains ≥70% UFA and <20% SFA

Downloaded from http://advances.sciencemag.org/ on February 2, 2021

Fig. 2. Dietary and adipose fat composition and parameters of local, systemic severity of CER pancreatitis in UFA- and SFA-fed ob/ob mice. (A1) Table comparing
the fatty acid composition of fat pad triglycerides (TG) and diets of mice given the SFA- and UFA-enriched diets. Body weights (A2), body fat (A3), and body fat as a per-
centage of body weight (A4) in the SFA- and UFA-fed mice. Serum amylase (B1) and lipase (B2) in control (CON) mice and after 24 hours of AP (CER). Local pancreatic in-
jury seen histologically (C1) and quantified as acinar necrosis (C2) as a percentage of total parenchymal area and percentage of acinar necrosis adjacent to the FN (shown
in yellow rectangles), termed as “% peri-fat acinar necrosis” (C3). Systemic injury measured as renal injury [TUNEL staining showing brown nuclei in (D1) and serum BUN
in (D2)], lung TUNEL positivity highlighted with arrows in (D3), and shown as number per high-power field (HPF) in (D4), along with shock (as measured by a drop in
carotid pulse distention) in (E1), serum calcium (E2), and survival curve (E3) of mice with CER AP. NS, not significant.

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 3 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

consistent with FDA recommendations, results in fat pad tri- deoxynucleotidyl transferase–mediated deoxyuridine triphosphate
glyceride to be composed of 18 ± 6% PA and 31 ± 14% LA, with nick end labeling) positivity, higher serum blood urea nitrogen
35 ± 6% oleic acid (OA), i.e., C18:1 in ob/ob mice (7). On feeding (BUN) levels (Fig. 2, D1 to D4), a greater decrease in carotid pulse
the special diets, the LA concentrations in the fat pads of the UFA- distention (consistent with shock), and SAP-associated hypocalcemia
and SFA-fed ob/ob mice correspondingly matched reports of those (55, 56) (Fig. 2, E1 and E2), resulting in 20% survival (P < 0.02 versus
in human adipose tissue from Japan [≈40% (25)] and the United 90% in the SFA group; Fig. 2E3). The findings of SAP were replicated
States [5% (26)]. By 8 to 14 weeks, the mice averaged 45.5 ± 0.5 g in in UFA-fed C57BL6 mice with diet-induced obesity (DIO), but not
both groups. CER AP reduced survival to 10% in the UFA group by the normal chow-fed mice (30.8 ± 0.7 g), which had mild pancreatitis
day 3 versus 90% in the SFA group (P < 0.02; fig. S6). We then sim- (fig. S7, A to F) (27). However, we did not use DIO as the primary
ulated the lower cutoff BMI (≤25) associated with SAP in countries model since the time to weight gain (>5 months) was prolonged.
with a higher %UFA intake. AP was initiated in UFA-fed mice
weighing 20 to 30% less and having 35 to 40% less adipose tissue Preferential lipolysis of unsaturated long-chain triglycerides
(Fig. 2, A2 to A4). AP increased serum amylase and lipase similarly worsens inflammation and systemic injury
in both groups (Fig. 2, B1 and B2) but was worse in the leaner UFA It has recently been shown that PNLIP mediates FN and severity of
group. The SFA group has lesser pancreatic necrosis (5.8 ± 0.8 versus AP (7). To understand the worse outcomes in UFA-fed mice, we
16.8 ± 4.7%, P = 0.024; Fig. 2, C1 and C2) especially bordering FN, compared the necrosed gonadal fat pads in both groups (Fig. 3A).
termed peri-fat acinar necrosis (2.7 ± 0.4 versus 6.9 ± 1.6%, P = 0.03; Grossly, the SFA group has reduced FN; however, the pancreatic
green outline, Fig. 2, C1 and C3). Consistent with SAP (53, 54), the amylase, lipase activity, and PNLIP protein (6- to 14-fold control)

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


UFA-fed mice had greater lung and renal tubular TUNEL (terminal had a large increase in both groups (Fig. 3, B1 and B2, and fig. S8A).

Fig. 3. Effect of visceral fat composition on its lipolysis and associated inflammatory response. (A) Gross appearance of the FN in SFA-fed (top) and UFA-fed mice
with pancreatitis. (B1) PNLIP, IKB-, and -tubulin (-Tub) amounts on Western blotting (B2) in the fat pads of SFA- or UFA-fed control (Con) mice or those with CER
pancreatitis (CER). Cytokine mRNA levels in fat pads (C1) shown as an increase over control levels or serum levels of IL-6 (C2), MCP-1 (C3), or TNF- (C4) in control mice or
those with CER pancreatitis after they had been fed with UFA- or SFA-enriched diets. Serum NEFA showing OA (D1), LA (D2), total serum UFA (C3), total serum NEFA (D4),
PA (D5), palmitoleic acid (D6), total serum SFA (D7), and serum SFA as a percentage of total NEFA (D8) in control mice or those with CER pancreatitis after they had been
fed with UFA- or SFA-enriched diets. Photo credit: Krutika Patel, Mayo Clinic, AZ.

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 4 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

The UFA mice had a larger cytokine mRNA increase in the fat pads of these in the visceral triglyceride, serum at baseline. The propor-
during AP and interleukin-6 (IL-6), monocyte chemoattractant tion of SFA in general went down with AP (Fig. 3D8). This general-
protein-1 (MCP-1), and tumor necrosis factor– (TNF-) proteins ized reduction in SFA release suggested that SFAs in triglyceride are
in the sera (Fig. 3, C1 to C4), which correlated with lower IB- an unfavorable substrate for PNLIP.
(42 ± 15% of controls, P < 0.05) in the necrosed UFA fat pads We next directly investigated whether visceral triglyceride
(Fig. 3B2). composition, irrespective of obesity or genetic background, affects
Since NEFA mediates cytokine increase (7), we measured serum its lipolysis and consequent severity during AP. For this experiment,
NEFA to understand the underlying mechanism. The UFA group the triglycerides of LA (C18:2), glyceryl trilinoleate (GTL), or PA
with AP had higher serum NEFA increase, especially UFAs, includ- (C16:0), i.e., glyceryl tripalmitate (GTP), were intraperitoneally giv-
ing C18:1 (OA) and C18:2 (LA) (Fig. 3, D1 to D4). This pattern has en to lean mice with a genetically dissimilar background than the
been noted during SAP-induced organ failure (7, 28, 53, 54) in both C57Bl6 mice, i.e., lean CD-1 mice. Two mechanistically distinct AP
rodents (5, 6, 27) and humans (4). However, inexplicably, the in- models—i.e., CER and IL12/18, both of which are mild in lean ro-
crease in C18:1, C18:2, and C16:1 (Fig. 3, D1, D2, and D6) was muted dents (27, 57)—were then induced (Fig. 4 and fig. S9).
in the SFA-fed mice, despite C18:1 being equal and C16:1 being Mice given GTL or GTP alone exhibited normal behavior, with
higher in the fat pads of the SFA group (Fig. 2A1 and fig. S5). Simi- no change in physiologic or biochemical parameters. During AP,
larly, C16:0 and SFA overall (Fig. 3, D5 and D7, and fig. S8B) in- neither GTL nor GTP affected the initial serum amylase or lipase
creased more in the UFA group, despite the SFA group having more increase (Fig. 4, A1 and A2, and fig. S9, A1 and A2), which is similar

Downloaded from http://advances.sciencemag.org/ on February 2, 2021

Fig. 4. Effect of intraperitoneal GTL or GTP on IL12,18-induced pancreatitis in lean CD-1 mice. Serum amylase (A1), lipase (A2), and trends and survival (A3) during
IL12/18 pancreatitis alone (IL12,18), with GTL (IL12,18 + GTL) or GTP (IL12,18 + GTP). Note similar amylase and lipase at day 2 in all groups and progressive worsening in
the GTL group, which has reduced survival. (B1 to B3) Representative H&E-stained images of the pancreas of mice belonging to the groups mentioned below the image.
Note the increased necrosis at the periphery of the lobules in IL12,18 + GTL group (black arrows). Box plots showing serum glycerol (C1) and NEFA (C2) concentrations in
control (CON) mice and in the different pancreatitis groups. (D1 to D3) Representative TUNEL-stained images of the kidneys of mice belonging to the groups mentioned
below the image. Note the increased positivity in the tubules of the IL12,18 + GTL group (black arrows), which also has a significantly higher BUN than other groups on
ANOVA (*) (D4). Box plots of the serum resistin (E1), IL-6(E2), TNF- (E3), and MCP-1(E4) control (CON) mice and in the different pancreatitis groups. * indicates significant-
ly higher than control on ANOVA, and # indicates a significant reduction compared to the IL12,18 + GTL group.

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 5 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

to ob/ob mice given UFA or SFA diets (Fig. 2, B1 and B2). The AP reduced lipolysis over 15 min. For example, LLP generated <30% LA,
outcomes also paralleled the UFA and SFA groups (Figs. 2 and 3) and PLP generated <9% LA versus GTL (Fig. 5B1). Both linoleate
with 0% survival in the GTL groups versus 80 to 90% in the GTP (4.3 ± 1 M) and oleate (4.4 ± 1.2 M) generation were markedly
groups (P < 0.01; Fig. 4A3 and fig. S9A3). The GTL group had worse reduced on LOP. This was paralleled by reductions in m and Cai
pancreatic necrosis (Fig. 4, B1 to B3, and fig. S9, B1 and B2), pre- increase (Fig. 5, B2 and B3).
dominantly at the periphery of the lobules exposed to the lipolytically
generated LA, resembling peri-fat acinar necrosis (Fig. 2, C1 to C3). Triglyceride saturation makes its lipolysis by PNLIP
The higher serum glycerol and corresponding NEFA in the GTL energetically and structurally unfavorable
groups with AP (Fig. 4, C1 and C2, and fig. S9, C1 to C3) supported We then studied the interaction and hydrolysis of GTL, LLP, and
preferential unsaturated triglyceride lipolysis. Serum BUN elevations, LOP focusing solely on triglyceride hydrolysis by human PNLIP in
renal tubular injury (Fig. 4, D1 to D4, and fig. S9, D1 and D2), lung phosphate-buffered saline (PBS; pH 7.4, 37°C, 150 mM Na). The
injury (fig. S10, A and B), and profound hypotension noted as shock hydrolysis, i.e., GTL > LLP > LOP, paralleled those in the acinar cell
(fig. S10C) were only noted in the GTL groups with AP. Consistent media (Fig. 5C1). We thus studied their interaction with PNLIP using
with the small adipose tissue mass and visceral FN in lean mice (fig. isothermal titration calorimetry (ITC).
S11, A1 and A2), there was no increase in serum resistin (Fig. 4E1 A single injection of human PNLIP (0.7 nmol/s) into a stable,
and fig. S9E1) and little or no increase in serum lactate dehydrogenase sonicated, stirred, and 100 M suspension of these triglycerides in
(LDH) (fig. S11, B and E). However, consistent with NEFA-driven PBS (at 300 s; fig. S13A) caused an endothermic interaction with a
inflammation (7), serum IL-6, TNF-, and MCP-1 were higher in magnitude paralleling lipolysis, i.e., GTL > LLP > LOP, the enthalpy of

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


both AP models with GTL (Fig. 4, E2 to E4, and fig. S9, E2 to E4), which paralleled the raw heat data (Fig. 5, C2 and C3, and fig. S13B).
perhaps due to the systemic lipotoxicity of LA noted as higher The enzyme kinetics studied with multiple-injection experiments
serum damage-associated molecular patterns (DAMPs), i.e., double-­ (fig. S13E) mirrored the above findings with the reaction rate (V;
stranded DNA (dsDNA) and histone-complexed DNA fragments micromolars per second) of GTL > LLP > LOP (fig. S13, F and G).
in the GTL groups with pancreatitis (fig. S11, C, D, F, and G). These The calculated kinetic parameters (fig. S13G) for GTL hydrolysis are
findings suggested that unsaturated visceral triglyceride is hydrolyzed similar to those previously published (60) and are more favorable
more than saturated triglyceride and worsens systemic inflammation than for LLP or LOP. Addition of orlistat (50 M) to PNLIP signifi-
and organ failure, consistent with what we note epidemiologically cantly reduced the H and Vmax in both types of injections (fig. S13,
(Fig. 1). We thus went on to study the mechanistic basis of this C, D, H, and I), supporting the relevance of the parameters to
in more detail. lipolysis. The above experiments thus showed that increasing satu-
ration makes the interaction of unsaturated triglycerides with PNLIP
Saturation reduces long-chain triglyceride lipolysis by energetically unfavorable, resulting in reduced lipolysis.
pancreatic lipases To verify the experimental results in an independent unbiased
To understand how visceral triglyceride composition influences manner, we undertook docking simulations using the open lid con-
AP severity, we simulated the in vivo AP-associated lipase leak into formation of the human PNLIP-procolipase complex (1LPA) (61)
fat using an in vitro system (Fig. 3, B1 and B2) (6, 7, 27). Acini release and studied access of the relevant triglycerides into the PNLIP ligand-­
pancreatic enzymes into the surrounding medium under the basal binding domain (LBD) via the oxyanion hole. Ser152, Asp176, and
state in vitro (58). Thus, we measured the hydrolysis of triglycerides His263 compose the catalytic triad of human PNLIP and are critical
added to the medium and biological responses of the NEFA generated. in fatty acid liberation from triglycerides. The closed conformation
Addition of pure triglycerides GTP, glyceryl trioleate (GTO; the (1LBP) (62) was not used since our goal was not to study molecular
triglyceride of OA, C18:1), or GTL (300 M each) to the stirred ac- dynamics of pancreatic lipase activation by interfaces.
inar medium in a quartz cuvette at 37°C resulted in 60 to 70% hy- Previous in silico studies (63) found that orlistat, a potent lipase
drolysis of GTL and GTO within 15 min but <10% of GTP (Fig. 5, inhibitor, docks to pancreatic lipase with a GlideScore of −6.90 kcal/mol
A1 and A2). Lipolysis of GTL and GTO, unlike GTP or GTLO (i.e., with root mean square deviation (RMSD) between 1.2 and 4.8 Å.
GTL + the lipase inhibitor orlistat at 50 M) which has <15% lipoly- This value served as a threshold to quantify strong binding. To verify
sis, also caused acinar mitochondrial depolarization (m) and cyto- this value, we docked orlistat to 1LPA with the induced fit protocol,
solic calcium (Cai) increase (Fig. 5, A3 and A4). Separately, on longer and a similar GlideScore value was produced. Orlistat docked with
incubation (6 hours), 80 to 100% of GTL was hydrolyzed to glycerol a distance of 3.74 Å between the catalytic serine and the -lactone
(fig. S12A) versus 20 to 30% of GTP even at a 3× molar excess. GTL ring that inhibits PNLIP hydrolysis. The induced fit protocol docked
hydrolysis caused cytochrome c leakage, decreased ATP (adenosine GTL into the 1LPA LBD with a GlideScore of −7.16 and with a distance
5′-triphosphate) levels, and increased LDH leakage and propidium of 4.04 Å between the hydroxyl group of Ser152 and the carbonyl C
iodide uptake (fig. S12, B to E). These studies showed that triglyceride atom of the triglyceride’s glycerol backbone (Fig. 5D1). LLP docked
saturation reduces its lipolysis and consequent biological effects. with a GlideScore of −4.72 kcal/mol at a distance of 9.99 Å from
We then studied the effect of saturation on the hydrolysis of mixed Ser152, and LOP and GTP respectively produced GlideScores of −1.33
triglycerides in the acinar medium, since triglycerides in vivo are mostly and −1.58 while being 12.42 and 11.98 Å from the catalytic serine
mixed. On the basis of pancreatic lipase lacking stereoselectivity for (Fig. 5, D2 to D4). To verify the integrity of the docking simulation,
the sn-1/sn-3 position (59), we chose mixed triglycerides (each at the ligand present in the 1LPA crystal structure (61), dilauryl phos-
100 M) of LA, i.e., 1,2-dilinoleoyl-3-palmitoyl-rac-glycerol (LLP) phatidyl choline (DLPC), was removed and then docked with the
(LA-LA-PA), 1,3-dipalmitoyl-2-linoleoylglycerol (PLP) (PA-LA-PA), same induced fit docking protocol used for the three triglycerides.
and 1-palmitoyl-2-oleoyl-3-linoleoyl-rac-glycerol (LOP) (LA-OA-PA), DLPC docked with a GlideScore of −7.46 (fig. S14A) and at a dis-
and compared these to GTL (LA-LA-LA). Palmitate disproportionately tance of 3.59 Å from the hydroxyl group of the catalytic serine,

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 6 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


Fig. 5. Influence of triglyceride composition on their lipolytic interaction, lipolysis and resulting biological effects. Lipase activity (A1), NEFA generation (A2), cy-
tosolic calcium (A3), and mitochondrial depolarization (A4) of acini alone (control) or with 300 M GTL, GTL with 50 M orlistat (GTLO), GTP, or GTO. (B1 to B3) Effect of
substituting LA in GTL (red) with palmitate at Sn-3 (LLP; magenta) or at Sn-1 and Sn-3 (PLP; cyan) or oleate and palmitate at Sn-2 and Sn-3, respectively (LOP; light brown)
on NEFA generation over 15 min by acini (B1), acinar mitochondrial depolarization (B2), and cytosolic calcium increase (B3). (C1) Time course of NEFA generation from
GTL, LLP, and LOP (100 M) by PNLIP (12 nM). (C2 and C3) hPNLIP-triglyceride interaction, showing raw heat per second (Q/t) as a function of PNLIP/triglyceride ratio
(C3) and measured peaks (C2). (D1) GTL docked into 1LPA via induced fit protocol. PNLIP molecule (top) and zoomed in LBD (bottom) are shown with catalytic triad (dark
blue), distances, and GlideScores (kilocalories per mole). LA, OA, and PA are in red, cyan, and green, respectively, with receptor interaction surface in yellow and oxygen
atoms in orange. (D2 to D4) LLP, LOP, and GTP docked into 1LPA.

which is 0.39 Å from its location in the crystal structure (3.20 Å). lipolysis (64). Unlike LA and OA, there was no increase in circulating
The resolution of 1LPA is 3.04 Å, so the variance seen is within the cytokines or organ failure (BUN increase) induced by PA (Fig. 6,
margin of error. DLPC inhibited the lipolysis of GTL added simul- A and B). The median survival for LA-treated mice was 18 hours
taneously in a dose-dependent manner (fig. S14B). This inhibition by and for OA was 64 hours, while the PA group had no adverse out-
DLPC, along with the redocking closely recapitulating the crystallo- come over 3 days. Serum NEFA (which are predominantly albumin
graphic findings, thus validates the docking simulation. Overall, these bound) and unbound NEFA (uNEFA; Fig. 6, C and D) increased
studies show that long-chain SFAs like palmitate make a triglyceride’s significantly only in the LA and OA groups, along with levels of the
interaction with PNLIP structurally and energetically unfavorable, DAMP and dsDNA (Fig. 6E) and a drop in serum albumin consistent
thus reducing its hydrolysis. We lastly went on to study how the fatty with the hypoalbuminemia noted during experimental (fig. S15, A
acids generated may result in cell injury and consequent organ failure. to C) and clinical SAP (55). This suggested that, unlike PA, both LA
and OA with high uNEFA may directly injure cells, triggering DAMP
Double bonds increase monomeric NEFA concentrations, release and downstream inflammation. This would also be consistent
signaling, and resulting organ failure with the in vivo AP models (Figs. 3 and 4 and figs. S9 and S11, B to
We first compared the ability of NEFA to directly induce inflamma- G) where higher NEFA were associated with worse outcomes.
tion and organ failure. LA [0.3% body weight (28, 53, 54)], OA (7), We thus compared the ability of the most prevalent long-chain
or PA (0.5%) body weight was instilled into the peritoneal cavity to NEFA (C18:2, C18:1, and C16:0; Fig. 6, F1 to F3) to exist in a non-
simulate NEFA generated from visceral fat (2 to 10% body weight) micellar monomeric form in aqueous media. On ITC at 37°C, we

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 7 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


Fig. 6. Effects of NEFA unsaturation on their in vivo effects and unbound monomeric behavior. Serum MCP-1 (A), BUN (B), NEFA (C), uNEFA (D), and dsDNA (E) after
intraperitoneally administering C16:0 (green), C18:1 (blue), and C18:2 (red) as mentioned below the x axis. Values are at necropsy if moribund or euthanasia at 72 hours.
(F1) ITC thermograms of injecting NEFA or solvent (0.34% DMSO for C16:0 and PBS for C18:1 or C18:2) at concentrations mentioned below the NEFA into PBS (pH 7.4) at
37°C, and corresponding enthalpograms (F2) and CMCs (F3) shown as shaded rectangles in the areas where they intersect the x axis. Effect of exogenous albumin (final
concentrations of 0.5%) on the cytosolic calcium (Cai) (G) and mitochondrial depolarization (m) (H and I) induced by 100 M of the indicated NEFA added at 100 s. (J and
K) Comparison of the dose responses of increase in mitochondrial depolarization (m) after 60 s of addition of different concentration of C18:1 (blue) or C18:2 (red) in
acini (J) and HEK293 cells (K). * indicates significant difference (P < 0.05) from the previous lower concentration by paired t test. # indicates a significant difference between
C18:2 and C18:1 for that specific concentration on t test.

noted the critical micellar concentration (CMC) and therefore the caused a larger increase in Cai (Fig. 6G) and m (Fig. 6H) than
aqueous monomeric NEFA concentrations to increase with the number 100 M OA (above CMC). PA caused no change in these. A total of
of double bonds (Fig. 6, F1 to F3). The CMCs of PA (C16:0), OA 0.5% albumin (Alb) completely aborted the Cai increase by both LA
(C18:1), and LA (C18:2) were respectively <8, ≈40, and ≈160 M, and OA and prevented m from progressing (Fig. 6, H and I). This
which paralleled the uNEFA levels in vivo (Fig. 6D). The calorimetric effect of albumin proves that the Cai and m increases were from
results were then validated using ultracentrifugation. Both of these the uNEFA. It also shows that Cai and m changes result from
methods could be performed at room temperature (23°C; fig. S16) distinct mechanisms of these uNEFA. The sustained m elevation,
and showed that the nonmicellar NEFA concentrations in the in- without progression, despite albumin (Fig. 6, H and I) signifying
franatents of 500 M NEFA in PBS (pH 7.4), spun at 105g for 1 hour irreversible uNEFA toxicity, is distinct from the complete reversal
(fig. S16, A and B), were similar to the CMCs on calorimetry (fig. of Cai by albumin (Fig. 6G) and explains the cell death noted from
S16, C and D) and those shown previously using diphenyl hexatriene LA or OA in previous studies (6). Therefore, the greater Cai and m
fluorescence spectroscopy (65). Thus, the CMC noted on calorimet- induced by LA than OA at 100 M (Fig. 6, G and H) may be due to
rically at 37°C (Fig. 6, F1 to F3) are accurate. LA’s higher CMC (Fig. 6F3) and thus explains the higher uNEFA
The role of uNEFA in causing biological effects was then studied levels and shorter survival noted in LA-treated mice. We tested this
in cells. Addition of 100 M LA (below CMC) to pancreatic acini by measuring the dose response of increase in m over baseline

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 8 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

(m) 60 s after the addition of LA or OA to acini (Fig. 6J) or human in aqueous media, unlike saturated NEFA, resulting in injurious sig-
embryonic kidney (HEK) 293 cells (Fig. 6K) to represent the naling, lipotoxic inflammation, and organ failure. This can potentially
pancreatic and kidney injury in vivo (Fig. 4, D1 to D4, and fig. S9, explain why higher dietary UFAs may result in worse AP in leaner
D1 and D2). Consistent with OA’s CMC (≈40 M; Fig. 6F3), m animals and humans with lower BMIs compared to the more obese
increased over baseline at 50 M OA (*) and then plateaued (Fig. 6, ones who consume a diet with higher proportions of saturated fat
J and K). Similarly, LA’s m increased till 100 M and ceased at (Fig. 7), resulting in the obesity paradox.
200 M, consistent with its CMC of ≈160 M. While LA’s m We note that the obesity paradox in pancreatitis (10) holds true
equaled OA’s at 50 M, it was more than OA’s (#) at concentrations when the data are grouped and analyzed by BMIs using the World
of ≥100 M. LA and OA (60 M), unlike PA, also reduced endothelial Health Organization (WHO) cutoffs (67) relevant to the countries
barrier integrity (fig. S15, D and E), potentially explaining the hypo- from which the study originated (Fig. 1), including those that use
tension (Fig. 2E1 and fig. S10C) hypothesized to be from vascular leak BMI cutoffs of ≤25, which have a lower SFA and higher %UFA con-
during SAP (66), along with explaining the hypoalbuminemia (fig. S15, sumption in diet (Fig. 1, B and C). Socioeconomic and quality-of-­
A to C) (55) in severe AP, and the uNEFA increase noted (Fig. 6D). care issues, age, sex, and etiology of AP are unlikely to have influenced
Overall, these studies cumulatively validate that double bonds in- our findings since the rate of SAP and mortality were the same in
crease monomeric long-chain NEFA concentrations and signaling the >30 versus ≤25 BMI cutoff groups. We note that the SAP rates
in an aqueous environment, thus enhancing their lipotoxicity. and %UFA intake have a moderate correlation (fig. S3B), and on
meta-regression, %UFA intake explains 33% of the heterogeneity in
SAP rates. Thus, other factors such as male sex, genetic background,

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


DISCUSSION and comorbidities that are not accounted for in the studies may
Here, we find that a higher proportion of dietary unsaturated fat can contribute to the remaining heterogeneity in SAP rates.
worsen AP outcomes at a lower adiposity than seen in individuals The difference in BMI cutoffs set by the WHO is commonly
with a higher proportion of saturated fats in their diet. We show attributed to a higher percentage fat per body mass in Asian popu-
that the higher likelihood of SAP associates with a higher unsaturated lations compared to Caucasian populations (68). However, this is
triglyceride content in visceral fat. This occurs because the presence brought into question since visceral adiposity (3) that undergoes
of SFAs in triglycerides makes the interaction of the substrate with visceral FN during SAP (69) differs by ethnic groups (70) and in
PNLIP structurally and energetically unfavorable. Moreover, the un- women versus men (71). We note that all visceral fat is not the same,
saturated NEFA generated by lipolysis can exist as monomers (65) since a higher proportion of diet-derived UFAs like LA make smaller

Fig. 7. Schematic summarizing how dietary fat composition affects visceral fat necrosis and causes the obesity paradox. The impact of consuming a Western diet en-
riched in saturated fat from dairy and red meat (left) or one enriched in unsaturated fat from vegetable oil and fish (right) are shown. In the event of AP, the relatively more
saturated adipocyte triglyceride shown in blue ( , left) or unsaturated triglyceride shown in red ( , right) is exposed to pancreatic lipase, principal among which is PNLIP
(zoomed circles). Despite being lesser in amount, the more unsaturated triglyceride is hydrolyzed more into the lipotoxic NEFA ( ), which are stable as monomers at
higher concentrations than saturated NEFA, that form micelles ( ) at lower concentrations. This results in worse systemic inflammation, injury, and organ failure in
those with a higher unsaturated fat consumption, despite having less adiposity. This pathophysiology can explain the obesity paradox.

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 9 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

amounts of visceral fat more prone to lipolysis (Figs. 2 and 3), gen- mirror the initial interaction. Previous studies suggest that this may
erating higher concentrations of monomeric NEFA (Fig. 6), thus be due to binding of the generated LA to aromatic residues on PNLIP
worsening outcomes in leaner populations (Fig. 1). The reverse holds and further increasing its activity (75).
true for SFAs, and this is relevant to studies from Western countries In summary, diet-induced visceral fat unsaturation increases
(72) that show AP severity to be independent of intra-abdominal fat lipolytic generation of unsaturated monomeric NEFA that cause cell
amounts even when this fat is above the range in studies from Asian injury, systemic inflammation, and organ failure. Long-chain SFAs
countries (71). This is exemplified by the six studies reporting a like palmitate, however, interfere with this lipolysis, generating lower
BMI of >30 to not be associated with severe AP—all of which came amounts of monomeric NEFA, making pancreatitis milder despite
from countries with <40% UFA as dietary intake (Fig. 1C). excess adiposity, thus explaining the obesity paradox (Fig. 7). The
Our epidemiologic studies are limited by being retrospective, not FDA guidelines to reduce saturated and increase unsaturated fat intake
involving dietary history, visceral fat sampling, or genetic studies, (https://www.fda.gov/files/food/published/Food-Labeling-Guide-
and by using nationwide dietary and economic data. We thus verified %28PDF%29.pdf, appendix F) (26, 52) could therefore potentially
the conclusions in mice, cell, and pure enzyme models and physical contribute to worsening systemic inflammation and organ failure.
chemistry. The use of two different strains of mice (i.e., C57bl/6 and
CD-1) and two modes of increasing visceral triglyceride (i.e., obesity
and intraperitoneal injection of triglyceride) experimentally support METHODS
that acute triglyceride lipolysis is crucial in the pathogenesis of organ Human data analysis
failure and weakens the argument in favor of the genetic or chronic Literature search

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


effects of obesity such as insulin resistance and atherosclerosis in To explore the impact of BMI on AP severity, we conducted a liter-
explaining these outcomes. The deleterious effects that we note with ature review by searching the PubMed database from inception to
GTL are also seen with the triglyceride of OA but not with GTP during January 2017). The terms “BMI” or “obese” and “acute pancreatitis”
pancreatitis (5). These along with the deleterious effect of monomeric as well as “severe” or “severity” were used. A total of 118 studies
unsaturated NEFA (Fig. 6), lipase (16–18), and NEFA elevation were retrieved, as shown (fig. S2). Seventy-nine studies were excluded,
(19, 20, 23), being associated with worse outcomes in other diseases as they were not related to the subject of this review, as were 12 studies
states including COVID-19 (22, 24), perhaps support the general that were reviews/meta-analyses/hypotheses. The remaining studies
relevance of dietary and visceral fat unsaturation in causing the were extensively reviewed for eligibility criteria, which were as fol-
obesity paradox (11–15). A palmitate-enriched diet in ob/ob mice lows: (i) clear BMI cutoff to define obesity, (ii) clear report of the
helped avoid the confounding effects of maldigestion of a dietary incidence of mild AP (MAP) and SAP in obese versus nonobese
saturated triglyceride by pancreatic lipases, which we note as reduced groups, and (iii) clear and satisfactory definitions of MAP and SAP.
lipolysis of GTP, LLP, and PLP (Fig. 5, A2 and B1). We used PNLIP Twenty-seven studies met our eligibility criteria and were therefore
since recent studies show that adipocyte triglyceride lipase, PNLIPRP2, included in this review (fig. S1). These are shown in different colors
and carboxyl ester lipase are unlikely to mediate lipotoxic systemic in Fig. 1A for descriptive purposes. The studies were then catego-
inflammation (7, 53). Moreover, the similar lipolysis pattern in acinar rized into those using a BMI of 30 as a cutoff (n = 20, the one with a
media, which contains all three lipases (Fig. 5, A1 to A4 and B1 to cutoff of >29 was included here) and those that used a cutoff BMI
B3) and PNLIP (Fig. 5, C1 to C3), lends credence of the concept to of ≤25 (n = 7; with text and country in pink) to define obesity’s
other lipases. association with SAP. The dietary fat consumption in the countries
Stearoyl-CoA (coenzyme A) desaturase-1(SCD-1) (73) is likely from which these papers originated was then extracted as described
responsible for converting the dietary palmitate (C16:0) to palmi- below and compared (fig. S1). The incidence of MAP and SAP in
toleate (C16:1), which comprised 17% of the visceral triglyceride of both obese and nonobese patients was extracted for the purpose of
SFA mice (Fig. 2A1 and fig. S5), despite palmitoleate being absent meta-analysis. Total number of patients was divided into MAP and
in the diet. SCD-1 is highly expressed in adipose tissue and can put SAP and then into those that were obese versus nonobese. In cases
a double bond in stearoyl-CoA or palmitoyl-CoA, thus forming where this was not explicitly clear, the communicating author was
oleate (C18:1) and palmitoleate from the palmitate in the diet. This contacted (30, 45), and the numbers so provided were included in
may also explain the similar amounts of C18:1 noted in the visceral the study. Papers that did not have adequate data or for reasons
triglyceride of the UFA- and SFA-fed mice. Despite the potential in- mentioned in the last column of fig. S1 and detailed in fig. S2 were
fluence of SCD-1 on our hypothesis, the similarity of dietary and vis- excluded from the meta-analysis (n = 7). When more than one BMI
ceral fat composition that we note in mice, the previous studies note in cutoff was mentioned, the BMI used was the one at which the SAP
humans (26), along with the lack of known pathways to saturate C18:1, risk increased.
and our mechanistic data cumulatively support our conclusions. Statistical analysis
An additional limitation may lie in our inability to resolve the The proportion of SAP in all pancreatitis was calculated and
lipolytic impact of stereochemical structure of the triglycerides (74), compared between obese and nonobese groups. Subgroup analysis
which we used in the racemic form rather than the enantiopure form. was performed on the basis of the definition of obesity used in the
While this remains beyond our scope, the previous findings that different studies included. Random effects model was used to calcu-
pancreatic lipase hydrolyzes triglyceride without any stereoselectivity late pooled OR with 95% CIs. Heterogeneity was assessed using
for the sn-1 or sn-3 position (59) suggest that the interference by the I2 measure and the Cochran Q statistic. Statistical analysis was
palmitate is independent of these locations on the glycerol backbone. performed using Comprehensive Meta-Analysis Software version
Last, the mechanisms underlying the exothermic changes (Fig. 5, C2 3.3.070 (Biostat, Englewood, NJ, USA). Publication bias was as-
and C3) following the initial interaction of PNLIP and triglycerides sessed for using Begg and Mazumdar’s test as well as Egger’s regres-
remain unclear. The relative magnitude of these, i.e., GTL > LLP > LOP, sion intercept.

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 10 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

Dietary fat consumption of country from which paper originated Committee of the Mayo Clinic (Scottsdale, AZ). Ex vivo studies were
The per capita per year dietary fat consumption of each country was col- done on pancreatic acini harvested from ICR mice (Charles River
lected from the FAO website (http://www.fao.org/faostat/en/#data/FBSH), Laboratories, Wilmington, MA). In vivo studies were done on
which is reported as kilograms per person per year. For purposes of genetically obese male ob/ob (B6.V-lepob/J; the Jackson laboratory,
simplicity, the chief sources of long-chain saturated fat (dairy, red meat, Bar Harbor, ME) mice given an experimental diet below or ICR mice.
and palm oil) were analyzed separately from those of unsaturated fat The mice were 7 to 14 weeks of age at the time of studies. There
(fish and vegetable oil). Fat from dietary sources were calculated were six to eight mice in each group.
by percentage weight (3.25% for whole milk, 20% from red meat,
and 10% for fish) unless the food source was directly fat (e.g., In vivo studies in mice
cream, butter, ghee, cheese, vegetable oil, or palm oil). Data from Experimental diets
the country from which each paper originated were included in the These were started immediately in ob/ob [B6.Cg-Lepob/J; Jax Stock
categories “BMI >30 not related to SAP,” “BMI >30 associated with #00632, the Jackson laboratory (Bar Harbor, ME)] or C57bl/6J (to
SAP,” and cutoff “BMI ≤25” (shown as blue, green with golden text induce DIO) after weaning (age of 4 to 5 weeks), with an intent to
or symbols, and pink in Fig. 1, respectively). Each country was rep- change visceral fat composition. The diets are detailed in fig. S4 and
resented once in the category for a contributed paper (shown in were given ad libitum. Normal chow [Purina 5053 diet, LabDiet
Fig. 1, B and C). Comparisons were done by grouping the BMI of (www.labsupplytx.com/wp-content/uploads/2012/10/5053.pdf),
>30 not related to SAP and BMI of >30 associated with SAP into a which contains 5.0% of which ≈70% is UFA] and water were fed to
grouped BMI of >30 category and by comparing this to the BMI of control mice ad libitum. Mice were weighed periodically (1 to 4 weeks,

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


≤25 category using a Mann-Whitney test. P < 0.05 was regarded depending on the rate of weight gain), and the body fat was mea-
as significant. sured using a Minispec Body Composition analyzer (Bruker Corpo-
ration, Billerica, MA) using nuclear magnetic resonance as previously
Reagents described (27).
Glyceryl trilinolein (GTL) and other triglycerides including triolein Experimental pancreatitis
(GTO), tripalmitin (GTP), and mixed triglycerides 1,2-dilinoleoyl-3-­ The models used were CER, and IL12 + IL-18 (IL12,18) induced
palmitoyl-rac-glycerol (LLP), 1-palmitoyl-2-oleoyl-3-linoleoyl-rac-glycerol pancreatitis in ob/ob mice, as detailed below. CER pancreatitis was
(LOP), 1,3-dipalmitoyl-2-linoleoylglycerol (PLP), 1,2-dioleoyl-3-­ also induced in C57bl/6 mice given the experimental diets or normal
palmitoyl-rac-glycerol (OOP), 1,3-dioleoyl-2-palmitoylglycerol chow after 28 weeks. CER pancreatitis was induced by hourly intra-
(OPO), 1,3-dipalmitoyl-2-oleoylglycerol (POP), linoleic acid (LA), peritoneal injections of CER (50 g/kg; Bachem AG, Bubendorf,
palmitic acid (PA), oleic acid (OA), dimethyl sulfoxide (DMSO), and Switzerland) × 12 doses on two consecutive days as described before
glycerol reagent were purchased from Sigma-Aldrich (St. Louis, MO). (27). IL12 (150 ng per dose per mouse; PeproTech) and IL18 (750 ng
CER (Bachem AG, Bubendorf, Switzerland), IL12 (PeproTech, Rocky per dose per mouse; R&D Systems) pancreatitis was induced by giving
Hill, NJ), IL18 (R&D Systems, Minneapolis, MN), PNLIP, IB-, IL12,18 as two doses 24 hours apart to lean ICR mice (10 to 14 weeks)
cytochrome c antibody (Santa Cruz Biotechnology, Dallas, TX), as described previously (57). Both agents were dissolved in PBS and
-tubulin [Developmental Studies Hybridoma Bank (DSHB), Uni- given intraperitoneally. When studying the effect of intraperitoneal
versity of Iowa, Iowa], Cox-IV antibody, and TO-PRO-3 iodide GTL (150 l) or GTP (150 mg) in these models, either agent was
(Thermo Fisher Scientific, Waltham, MA) were used. given 4 hours after the first CER injection or second IL12,18 injec-
tions. NEFA were administered into the peritoneal cavity of mice as
Use of lipids described previously (7, 22, 28).
GTL, other triglycerides, and fatty acids were sonicated into the media Carotid pulse distension (MouseOx Oximeter, STARR Life Sci-
in a two-step fashion to ensure that the triglyceride stays in solution ences, Pittsburgh, PA) was used as a noninvasive measure of blood
as described previously (53). Briefly, pure GTL, GTO, LLP, LOP, pressure. Mice were observed four times a day and were electively
and OOP were first directly sonicated into the PBS (concentration, sacrificed at the end of the experiment using carbon dioxide, unless
3 mM) using three 10-s pulses. This sonicate was promptly added to they became moribund prematurely. Terminal blood and tissue
the medium (final concentration, 300 M). The only triglycerides harvest was done if the animal was moribund and used in the analyses,
requiring a solvent (DMSO) were those with two or more PA chains unless specified for a different time point. Tissues were collected and
(PLP, GTP, and POP). These were prepared as follows: triglyceride preserved for mRNA (RNA later), activity (−80°C), histology and
stock (60 mM) was prepared in 100% DMSO by heating. These stocks TUNEL staining (formalin), and Luminex and biochemical assays
were sonicated into PBS (concentration, 3 mM and 5% DMSO). (serum) as described previously (6, 7, 28).
These sonicates were quickly added to the medium (final concen- Histology and TUNEL staining
tration, 300 M and 0.5% DMSO). The experimental endpoints Pancreas, kidney, and lung tissue were fixed with 10% neutral buff-
were not affected by this 0.1 to 0.5% DMSO in our system, as shown ered formalin and processed for embedding in paraffin. Sections
previously (6). (5 m) were used for hematoxylin and eosin (H&E) staining and for
TUNEL staining as described previously (5, 6, 27). Digital images of
Animal experiments section were captured with a digital microscope Axio Imager M2 or
All animals were acclimatized for a minimum of 2 days before use. Axio Observer Z1 (Zeiss, Oberkochen, Germany) with a 20× objec-
These were housed with a 12-hour light/dark cycle at room tem- tive with 10 random images per section and quantified as number of
perature, fed normal laboratory chow (ICR mice) or an experimental TUNEL-positive nuclei/high field of lung tissue. Pancreatic necrosis
diet (see below), and were allowed to drink ad libitum. All experi- was quantified on whole-pancreas H&E-stained sections after being
ments were approved by the Institutional Animal Care and Use examined by a trained morphologist blinded to the sample. Briefly,

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 11 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

all pancreatic parenchymal area was imaged using the PathScan pyruvate) as previously described (6, 7, 28). Triglycerides or NEFA
Enabler IV slide scanner (Meyer Instruments, Houston, TX), and were added after 2 min of stirring at a 1× concentration to the
images were evaluated for total acinar area, total acinar necrosis, and stirred cell suspensions in a quartz cuvette at 37°C, and changes in
acinar necrosis in direct proximity to the necrosed fat, i.e., peri-fat intracellular calcium concentrations were determined by alternate
acinar necrosis as described previously (6, 27) in pixels for each excitation at 340 and 380 nm, measuring emission at 510 nm. Mito-
pancreas. Percentage necrosis was reported as a percentage of the chondrial inner membrane potential values (m) were determined
total acinar area for each pancreas. by excitation at 490 nm and alternate measuring emission at 530 and
In vitro cell studies 590 nm using the F2100 Hitachi Fluorescence Spectrophotometer.
All data shown are from a three to five independent experiments. The data were collected every 10 s. The first 2 min of data were
Use of lipids averaged and subtracted, and the net increase was plotted as a func-
Triglycerides (e.g., GTL, LLP, LOP, and GTO) or NEFA were soni- tion of time or NEFA concentrations. In some studies, excess (i.e.,
cated at 10× concentrations into the medium, and the only solvent 0.5%) fatty acid–free bovine serum albumin was added to neutralize
used was 5% DMSO (as a 10× stock) for GTP and PLP or for palmitate, and thus determine the changes attributable to the uNEFA.
since these otherwise were insoluble and precipitated out in the
medium. Solvents were otherwise avoided, since lipolysis in vivo takes Assays
place without solvents. These agents were added at a 1× concentration. Biochemical assays
Pancreatic acinar harvest and use The assays (amylase, lipase, BUN, and calcium; Pointe Scientific,
Eight- to 12-week-old ICR mice (Charles River Laboratories, Wilmington, Canton, MI), DNA damage: Quant-iT PicoGreen dsDNA reagent

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


MA) were euthanized, and pancreatic acini were harvested as de- (Life Technologies, Carlsbad, CA), LDH leakage using the Cytotoxicity
scribed (7, 28). Detection Kit (Roche, Mannheim, Germany), intracellular ATP mea-
Cell lines and use surement using ATP assay kit (CellTiter-Glo 2.0 Assay, Promega,
HEK293 cells were maintained in Eagle’s minimum essential media Madison, USA), glycerol using free glycerol reagent, and standard
[American Type Culture Collection (ATCC), Manassas, VA] sup- (Sigma-Aldrich, St. Louis, MO) and colorimetric total free fatty acids
plemented with 10% fetal bovine serum (ATCC, Manassas, VA) using NEFA kit (FUJIFILM Wako Diagnostics, Mountain View, CA)
and 1% penicillin/streptomycin (Life Technologies, Carlsbad, CA). were done as per the manufacturer’s protocol (5, 6, 27).
All cell cultures were maintained in a humidified 5% CO2 atmosphere uNEFA measurement by fluorescence
in air at 37°C. Human endothelial cells (HUV-EC-C) were main- uNEFAs were measured by the emission at 550/457 nm upon exci-
tained in Kaighn’s modification of Ham’s F-12 medium (F-12K, tation at 375 nm and calculated as described in the ADIFAB2 kit
ATCC, Manassas, VA) supplemented with 10% fetal bovine serum (FFA Sciences LLC, CA, USA) protocol version 1.0 6-9-04. Standard
(ATCC, Manassas, VA), 1% penicillin/streptomycin (Life Technol- concentration versus fluorescence ratio curves were made with known
ogies, Carlsbad, CA), heparin (0.1 mg/ml; Millipore Sigma, St. Louis, concentrations of either >99.5% purity LA, OA, or PA (1, 2, 5, 10,
MO), and Corning endothelial cell growth supplement (50 g/ml; 20, 30, 50, and 100 M) in DMSO to have these all in the pure
Thermo Fisher Scientific). monomeric form. Then, the unknown (e.g., mouse serum) was mea-
TEER and dextran permeability assay sured and reported as uNEFA (micromolar concentrations), based
HUV-EC-C cells were grown as monolayers on a 0.4-m polyester on the fatty acid standard curve.
Transwell permeable membrane (Corning, NY) precoated with Cytokine/chemokine assays
type IV collagen and then used for trans-endothelial electrical resistance Serum cytokine/chemokine protein levels were assayed with a
(TEER) measurement and permeability tracer flux assay. Before MILLIPLEX MAP Mouse Adipokine Magnetic Bead Panel (Millipore,
studies, cells were suspended in Hepes buffer (pH 7.4) (20 mM Burlington, MA) according to the manufacturer’s recommendations
Hepes, 120 mM NaCl, 5 mM KCl, 1 mM MgCl2, 10 mM glucose, on a Luminex 200 System (Life Technologies) and analyzed using
10 mM sodium pyruvate, and 1 mM CaCl2), and exposed to NEFA the xPONENT software.
at the indicated concentration (60 M). TEER measurements were Triglyceride lipid composition and NEFA analysis
made using EVOM2 Epithelial Voltohmmeter with an STX2 elec- These were done at the Hormone Assay and Analytical Services Core
trode (World Precision Instruments, Sarasota, FL). Results were (Vanderbilt University Medical Center). For tissue triglyceride com-
presented as absolute values (Ω·cm2) after 2 hours of treatment. position, the Folch method (76) was used, and the lipids recovered
To evaluate the paracellular permeability of cultured HUV-EC-C in the chloroform phase were separated by thin-layer chromatography
cells, 10 kD of Dextran Alexa Fluor 647 (Thermo Fisher Scientific, using Silica Gel 60A plates. The triglyceride fraction was methylated
Waltham, MA) was added to the upper chamber, and the increase using the BF3/methanol method (77). These were analyzed using gas
in fluorescence was measured in the lower chamber 2 hours after chromatography (Agilent 7890A) using a capillary column (SP2380,
stimulation with NEFA. 0.25 mm by 30 m, 0.25-m film; Supelco, Bellefonte, PA) and helium
For in vitro calcium ion flux and mitochondrial depolarization as a carrier gas. Methyl esters of the fatty acids were identified by
studies, cells were loaded with fura-2AM (5 g/ml; Molecular Probes, comparing their retention times to those of known standard flame
Invitrogen) and 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimid- ionization detection. Internal lipid standards for quantification in-
azolylcarbocyanine iodide (JC-1, 5 g/ml; Enzo Life Sciences, cluded dipentadecanoyl phosphatidylcholine (C15:0), diheptadecanoin
Farmingdale, NY) at 37°C for 30 min in the media that they were (C17:0), trieicosenoin (C20:1), and cholesteryl eicosenoate (C20:1).
cultured or harvested in. After this, these were stored on ice till just Serum-free fatty acids were extracted as described previously (78) in
before use. Just before the experiment, the cells were washed and 30:70 heptane:isopropanol. This was acidified with 0.033 N H2SO4,
suspended in Hepes buffer (pH 7.4) (20 mM Hepes, 120 mM NaCl, and fatty acids were recovered in the heptane layer. Thin-layer
5 mM KCl, 1 mM MgCl2, 10 mM glucose, and 10 mM sodium chromatography and gas chromatography were done as described

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 12 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

above. Dipentadecanoic acid (C15:0) was added as an internal stan- and V values and summarized as means ± SEM from three indepen-
dard. UFA amounts were calculated by adding individual C16:1, C18:1, dent experiments.
C18:2, C18:3, C20:4, and C20:5 fatty acids. SFA amounts were calcu- From the raw data of continuous injection, enthalpy of the reac-
lated by adding individual C12:0, C14:0, C16:0, and C18:0 fatty acids. tion was calculated from the delta heat change per mole. Interaction
Western blotting of triglyceride and hPNLIP followed by hydrolysis of triglyceride was
Western blotting was performed as described previously (54). Briefly, graphically presented from three independent experiments.
cell lysate or fat pad lysate was prepared in radioimmunoprecipitation ITC for CMC measurement experiments
assay lysis buffer containing various protease inhibitors (complete We followed the method described previously (54). A total of 20 ali-
and free of EDTA; Roche, Pleasanton, CA). Protein concentration quots of degassed LA (0.7 mM, 2.5 l per injection from PBS, pH 7.4),
(1 mg/ml) of the lysate was adjusted, boiled (10 min) in Laemmli OA (0.34 mM, 2.0 l per injection from PBS, pH 7.4), and PA (0.34 mM
buffer, and run on (10 mg per lane) 4 to 20% gradient polyacrylamide in 0.34% DMSO, 2.0 l per injection from PBS, pH 7.4) were injected
gels. Then, proteins were transferred from the gel to polyvinylidene (10 M per injection for LA and 4 M per injection for both OA and
difluoride membrane (Merck KGaA, Darmstadt, Germany) and PA) from a rotating syringe (350 rpm) into the ITC sample chamber
blocked with 5% nonfat dry milk incubated with primary antibody containing PBS equilibrated at 37°C. Double-distilled water was used
of PNLIP (1:1000; Santa Cruz Biotechnology, Dallas, TX), IB- in a reference electrode chamber. The interval between two injections
(1:1000; Santa Cruz Biotechnology), -tubulin (1:500; DSHB, Uni- was kept at 200 s. The control experiment was performed by injecting
versity of Iowa, Iowa), cytochrome c (1:1000; Santa Cruz Biotech- PBS into PBS and DMSO (0.34%) into PBS.
nology), and Cox-IV (1:1000; Thermo Fisher Scientific, Waltham, MA) Ultracentrifugation studies

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


separately. Then, blots were probed with horseradish peroxidase– A total of 500 M C18:1, C18:2, and C18:3 were probe-sonicated
labeled corresponding secondary antibodies (1:10,000; Millipore, into PBS (pH 7.4) at room temperature. For C16:0 and C18:0, 150 mM
Billerica, MA). Band intensity was visualized by chemiluminescence stocks were prepared in pure DMSO, and these were then serially
using the Pierce ECL 2 Western Blotting Substrate Kit (Thermo Fisher sonicated and diluted down to 500 M (0.34% DMSO) in PBS (pH 7.4)
Scientific) and quantified by densitometry as described previously (79). at room temperature. A total of 4.8 ml of sample was loaded in
Isothermal titration calorimetry OptiSeal 4.9-ml tubes (Beckman Instruments Inc., Palo Alto, CA)
The hydrolysis of triglycerides by hPTL was carried out on a Nano in a NVT90 fixed-angle rotor (Beckman Instruments Inc., Palo Alto,
ITC (TA Instruments-Waters LLC, New Castle, DE) instrument and CA) and spun at 105g for 1 hour at room temperature in a Beckman
through Nano ITCRun Software v3.5.6. The experimental setup was Coulter Optima L-100 XP Ultracentrifuge (Beckman Coulter Inc.,
equipped with a computer-controlled micro-syringe injection device, IN, USA). The bottom of the tubes was then pierced with a 25-gauge
which allowed to inject small amounts of stock solution of even 0.1 l needle, and 1.0 ml of the liquid was removed. NEFA concentrations
to the sample chamber. Enzyme reaction was performed by two in these were measured using the colorimetric NEFA kit (FUJIFILM
methods: either incremental injection of substrate to enzyme or Wako Diagnostics, Mountain View, CA).
continuous injection of enzyme to substrate. Molecular docking
Incremental injection Protein Data Bank structure 1LPA (interfacial activation of the
A total of 10 aliquots of freshly prepared, degassed GTL or LLP or lipase-procolipase complex by mixed micelles revealed by x-ray
LOP (0.4 mM, 5 l per injection) were injected from a rotating crystallography) was downloaded from the RCSB protein data
syringe of speed 350 rpm into the ITC sample chamber containing bank (80) (rcsb.org) and then imported to the Schrödinger Maestro
hPNLIP (12 nM, ~500 U/liter activity) equilibrated at 37°C. The in- (Schrödinger Release 2019-4; Maestro, Schrödinger LLC, New York,
terval between two injections was 300 s. Similarly, individual tri- NY, 2019). Using the protein preparation wizard (Schrödinger
glyceride into PBS or PBS into hPNLIP was run as a control. To Release 2019-4: Protein Preparation Wizard; Epik, Schrödinger LLC,
confirm the specificity, LLL was injected into hPNLIP having orlistat New York, NY, 2016; Impact, Schrödinger LLC, New York, NY,
(50 M orlistat; Cayman Chemical Company, Ann Arbor, MI) in a 2016; Prime, Schrödinger LLC, New York, NY, 2019), 1LPA was pre-
similar process. processed at pH 7.4, and a restrained minimization was completed
Continuous injection by converging when the RMSD between iterations was less than
Degassed hPNLIP (45 l, 120 nM) was injected during 900 s from a 0.30 Å. The processed 1LPA structure was rendered in an aqueous
rotating syringe into the ITC sample chamber containing LLL or environment at pH 7.4 in the presence of 150 mM sodium ions and
LLP or LOP (0.1 mM) equilibrated at 37°C. The control experiment 150 mM chloride ions by using the Desmond System Builder
was performed by injecting PBS into hPNLIP. hPNLIP-mediated (Schrödinger Release 2019-4: Desmond Molecular Dynamics System,
hydrolysis of LLL was confirmed using hPNLIP with orlistat in the D. E. Shaw Research, New York, NY, 2019; Maestro-Desmond
same experimental setup. Interoperability Tools, Schrödinger, New York, NY, 2019) within
Calculation Schrödinger Maestro to mimic the physiological environment that
The ITC raw data were analyzed using the NanoAnalyze v3.10.0 the enzyme functions in.
software. Maximum velocity at saturating substrate concentration All structures to be docked (GTL, GTP, LLP, LOP, and DLPC)
(Vmax) was determined. Michaelis constant (Km), the measure of were downloaded as SDF files from PubChem (81) and imported to
dissociation/affinity of enzyme-substrate complex, was calculated Schrödinger Maestro. The LigPrep (Schrödinger Release 2019-4:
from the substrate concentration at Vmax/2. Enzyme turnover rate LigPrep, Schrödinger LLC, New York, NY, 2019) feature was used
(Kcat) was calculated by Vmax/enzyme concentration. Substrate to to obtain the ionization states of the ligands at pH 7.4 and to gener-
product formation rate (V) was calculated by the equation [Kcat × ate tautomers of the ligands. An induced fit docking (Schrödinger
enzyme concentration × substrate concentration]/[Km + substrate Release 2019-4: Induced Fit Docking protocol; Glide, Schrödinger
concentration]. For each experiment, we calculated Vmax, Km, Kcat, LLC, New York, NY, 2016; Prime, Schrödinger LLC, New York,

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 13 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

NY, 2019) was performed by selecting the four triglycerides as the 11. K. Al-Tarrah, S. W. Jones, N. Moiemen, J. M. Lord, Potential role of adipose tissue and its
hormones in burns and critically iII patients. Burns, 259–266 (2020).
ligands to be docked and designating the 1,2-didodecanoyl-sn-glycero-­
12. G. C. Fonarow, P. Srikanthan, M. R. Costanzo, G. B. Cintron, M. Lopatin; ADHERE Scientific
3-phosphocholine ligand from the 1LPA crystal structure as the Advisory Committee and Investigators, An obesity paradox in acute heart failure:
centroid of the workspace ligand around which the receptor grid Analysis of body mass index and inhospital mortality for 108,927 patients in the Acute
was generated. The ligand diameter midpoint box was constructed Decompensated Heart Failure National Registry. Am. Heart J. 153, 74–81 (2007).
with dimensions of X, 10 Å; Y, 10 Å; and Z, 10 Å; while the receptor 13. P. S. Whiting, G. A. White-Dzuro, F. R. Avilucea, A. C. Dodd, N. Lakomkin, W. T. Obremskey,
C. A. Collinge, M. K. Sethi, Body mass index predicts perioperative complications
grid was generated with dimensions of X, 30 Å; Y, 30 Å; and Z, 30 Å. following orthopaedic trauma surgery: An ACS-NSQIP analysis. Eur. J. Trauma Emerg. Surg.
No constraints were imposed to ensure that the docking simulation was 43, 255–264 (2017).
unbiased. The induced fit docking used the OPLS3e force field (82). 14. M. Hartrumpf, R.-U. Kuehnel, J. M. Albes, The obesity paradox is still there: A risk analysis
GlideScore docking energies were recorded in kilocalories per mole. of over 15 000 cardiosurgical patients based on body mass index. Interact. Cardiovasc.
Thorac. Surg. 25, 18–24 (2017).
Statistical analysis and graphical representation
15. Y. Zhao, Z. Li, T. Yang, M. Wang, X. Xi, Is body mass index associated with outcomes
Independent variables for in vivo studies are shown in box plots in of mechanically ventilated adult patients in intensive critical units? A systematic review
which the mean (dotted line), median (solid line), and in vitro data and meta-analysis. PLOS ONE 13, e0198669 (2018).
are shown as bar graphs reported as means ± SEM. Line graphs were 16. A. Subramanian, V. Albert, B. Mishra, S. Sanoria, R. M. Pandey, Association between
used for continuous variables. All values are reported as means ± SD. the pancreatic enzyme level and organ failure in trauma patients. Trauma Mon. 21,
e20773 (2016).
Significance levels were evaluated at P < 0.05. Data for multiple groups 17. J. Manjuck, J. Zein, C. Carpati, M. Astiz, Clinical significance of increased lipase levels
were compared by one-way analysis of variance (ANOVA) versus on admission to the ICU. Chest 127, 246–250 (2005).
controls, and values significantly different from controls were shown 18. D. W. Rattner, Z. Y. Gu, G. J. Vlahakes, A. L. Warshaw, Hyperamylasemia after cardiac

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


as (*) or with the P value mentioned above the corresponding con- surgery. Incidence, significance, and management. Ann. Surg. 209, 279–283 (1989).
19. P. E. Cogo, G. Giordano, T. Badon, A. Orzali, L. J. I. Zimmermann, F. Zacchello, P. J. J. Sauer,
ditions. When comparing two groups, a t test or Mann-Whitney test
V. P. Carnielli, Simultaneous measurement of the rates of appearance of palmitic
was used, depending on the normality of distribution and shown as and linoleic acid in critically ill infants. Pediatr. Res. 41, 178–182 (1997).
(*) when significantly different; alternately, P values are shown in 20. K. Kosaka, K. Suzuki, M. Hayakawa, S. Sugiyama, T. Ozawa, Leukotoxin, a linoleate
the graph. Graphing was done using SigmaPlot 12.5 (Systat Software epoxide: Its implication in the late death of patients with extensive burns. Mol. Cell.
Inc., San Jose, CA). Biochem. 139, 141–148 (1994).
21. F. Liu, X. Long, B. Zhang, W. Zhang, X. Chen, Z. Zhang, ACE2 expression in pancreas may
cause pancreatic damage after SARS-CoV-2 infection. Clin. Gastroenterol. Hepatol. 18,
SUPPLEMENTARY MATERIALS 2128–2130.e2 (2020).
Supplementary material for this article is available at http://advances.sciencemag.org/cgi/ 22. B. El-Kurdi, B. Khatua, C. Rood, C. Snozek, R. Cartin-Ceba, V. P. Singh; Lipotoxicity in
content/full/7/5/eabd6449/DC1 COVID-19 Study Group, Mortality from coronavirus disease 2019 increases
with unsaturated fat and may be reduced by early calcium and albumin
View/request a protocol for this paper from Bio-protocol.
supplementation. Gastroenterology 159, 1015–1018.e4 (2020).
23. T. Thomas, D. Stefanoni, J. A. Reisz, T. Nemkov, L. Bertolone, R. O. Francis, K. E. Hudson,
REFERENCES AND NOTES J. C. Zimring, K. C. Hansen, E. A. Hod, S. L. Spitalnik, A. D’Alessandro, COVID-19 infection
1. I. C. Funnell, P. C. Bornman, S. P. Weakley, J. Terblanche, I. N. Marks, Obesity: An important alters kynurenine and fatty acid metabolism, correlating with IL-6 levels and renal status.
prognostic factor in acute pancreatitis. Br. J. Surg. 80, 484–486 (1993). JCI Insight 5, e140327 (2020).
2. R. C. Turner, R. McDermott, Clinical predictors of severe acute pancreatitis: Value-adding 24. P. Hegyi, Z. Szakács, M. Sahin-Tóth, Lipotoxicity and cytokine storm in severe acute
the view from the end of the bed. ANZ J. Surg. 84, 672–676 (2014). pancreatitis and COVID-19. Gastroenterology 159, 824–827 (2020).
3. O. Sadr-Azodi, N. Orsini, Å. Andrén-Sandberg, A. Wolk, Abdominal and total adiposity 25. C. Fujii, T. Kawai, K. Azuma, Y. Oguma, F. Katsukawa, H. Hirose, K. Tanaka, S. Meguro,
and the risk of acute pancreatitis: A population-based prospective cohort study. Am. H. Matsumoto, H. Itoh, Relationships between composition of major fatty acids and fat
J. Gastroenterol. 108, 133–139 (2013). distribution and insulin resistance in Japanese. J. Diabetes Res. 2017, 1567467 (2017).
4. S. Domschke, P. Malfertheiner, W. Uhl, M. Büchler, W. Domschke, Free fatty acids in serum 26. S. J. Guyenet, S. E. Carlson, Increase in adipose tissue linoleic acid of US adults in the last
of patients with acute necrotizing or edematous pancreatitis. Int. J. Pancreatol. 13, half century. Adv. Nutr. 6, 660–664 (2015).
105–110 (1993). 27. K. Patel, R. N. Trivedi, C. Durgampudi, P. Noel, R. A. Cline, J. P. DeLany, S. Navina,
5. P. Noel, K. Patel, C. Durgampudi, R. N. Trivedi, C. de Oliveira, M. D. Crowell, R. Pannala, V. P. Singh, Lipolysis of visceral adipocyte triglyceride by pancreatic lipases converts mild
K. Lee, R. Brand, J. Chennat, A. Slivka, G. I. Papachristou, A. Khalid, D. C. Whitcomb, acute pancreatitis to severe pancreatitis independent of necrosis and inflammation. Am.
J. P. De Lany, R. A. Cline, C. Acharya, D. Jaligama, F. M. Murad, D. Yadav, S. Navina, J. Pathol. 185, 808–819 (2015).
V. P. Singh, Peripancreatic fat necrosis worsens acute pancreatitis independent 28. B. Khatua, J. R. Yaron, B. El-Kurdi, S. Kostenko, G. I. Papachristou, V. P. Singh, Ringer’s
of pancreatic necrosis via unsaturated fatty acids increased in human pancreatic necrosis lactate prevents early organ failure by providing extracellular calcium. J. Clin. Med. 9, 263
collections. Gut 65, 100–111 (2016). (2020).
6. S. Navina, C. Acharya, J. P. De Lany, L. S. Orlichenko, C. J. Baty, S. S. Shiva, C. Durgampudi, 29. L. Hodson, C. M. Skeaff, B. A. Fielding, Fatty acid composition of adipose tissue and blood
J. M. Karlsson, K. Lee, K. T. Bae, A. Furlan, J. Behari, S. Liu, T. M. Hale, L. Nichols, in humans and its use as a biomarker of dietary intake. Prog. Lipid Res. 47, 348–380 (2008).
G. I. Papachristou, D. Yadav, V. P. Singh, Lipotoxicity causes multisystem organ failure 30. P. J. W. Lee, A. Bhatt, J. Holmes, A. Podugu, R. Lopez, M. Walsh, T. Stevens, Decreased severity
and exacerbates acute pancreatitis in obesity. Sci. Transl. Med. 3, 107ra110 (2011). in recurrent versus initial episodes of acute pancreatitis. Pancreas 44, 896–900 (2015).
7. C. de Oliveira, B. Khatua, P. Noel, S. Kostenko, A. Bag, B. Balakrishnan, K. S. Patel, 31. P. Mentula, M.-L. Kylänpää, E. Kemppainen, H. Repo, P. Puolakkainen, Early inflammatory
A. A. Guerra, M. N. Martinez, S. Trivedi, A. M. Cullough, D. M. Lam-Himlin, S. Navina, response in acute pancreatitis is little affected by body mass index. Scand.
D. O. Faigel, N. Fukami, R. Pannala, A. E. Phillips, G. I. Papachristou, E. E. Kershaw, J. Gastroenterol. 42, 1362–1368 (2007).
M. E. Lowe, V. P. Singh, Pancreatic triglyceride lipase mediates lipotoxic systemic 32. V. P. Deenadayalu, U. Blaut, J. L. Watkins, J. Barnett, M. Freeman, J. Geenen, M. Ryan,
inflammation. J. Clin. Invest. 130, 1931–1947 (2020). H. Parker, J. T. Frakes, E. L. Fogel, W. B. Silverman, K. S. Dua, G. Aliperti, P. Yakshe, M. Uzer,
8. R. B. Thandassery, S. Appasani, T. D. Yadav, U. Dutta, A. Indrajit, K. Singh, R. Kochhar, W. Jones, J. Goff, M. Temkit, G. A. Lehman, S. Sherman, Does obesity confer an increased
Implementation of the Asia-Pacific guidelines of obesity classification on the APACHE-O risk and/or more severe course of post-ERCP pancreatitis?: A retrospective, multicenter
scoring system and its role in the prediction of outcomes of acute pancreatitis: A study study. J. Clin. Gastroenterol. 42, 1103–1109 (2008).
from India. Dig. Dis. Sci. 59, 1316–1321 (2014). 33. S. Sawalhi, H. Al-Maramhy, A. I. Abdelrahman, S. E. G. Allah, S. Al-Jubori, Does
9. K. Y. Shin, W. S. Lee, D. W. Chung, J. Heo, M. K. Jung, W. Y. Tak, Y. O. Kweon, C. M. Cho, the presence of obesity and/or metabolic syndrome affect the course of acute
Influence of obesity on the severity and clinical outcome of acute pancreatitis. Gut Liver 5, pancreatitis?: A prospective study. Pancreas 43, 565–570 (2014).
335–339 (2011). 34. A. Brown, T. James-Stevenson, T. Dyson, D. Grunkenmeier, The panc 3 score: A rapid
10. R. Premkumar, A. R. Phillips, M. S. Petrov, J. A. Windsor, The clinical relevance of obesity and accurate test for predicting severity on presentation in acute pancreatitis. J. Clin.
in acute pancreatitis: Targeted systematic reviews. Pancreatology 15, 25–33 (2015). Gastroenterol. 41, 855–858 (2007).

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 14 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

35. K. A. Porter, P. A. Banks, Obesity as a predictor of severity in acute pancreatitis. Int. a Pinhão Coat (Araucaria angustifolia) extract rich in condensed tannin. Nutrients 7,
J. Pancreatol. 10, 247–252 (1991). 5601–5614 (2015).
36. C. J. Tsai, Is obesity a significant prognostic factor in acute pancreatitis? Dig. Dis. Sci. 43, 61. H. van Tilbeurgh, M.-P. Egloff, C. Martinez, N. Rugani, R. Verger, C. Cambillau, Interfacial
2251–2254 (1998). activation of the lipase-procolipase complex by mixed micelles revealed by x-ray
37. J. Martinez, J. Sánchez-Payá, J. M. Palazón, J. R. Aparicio, A. Picó, M. Pérez-Mateo, Obesity: crystallography. Nature 362, 814–820 (1993).
A prognostic factor of severity in acute pancreatitis. Pancreas 19, 15–20 (1999). 62. M.-P. Egloff, F. Marguet, G. Buono, R. Verger, C. Cambillau, H. van Tilbeurgh, The 2.46
38. C. D. Johnson, S. K. C. Toh, M. J. Campbell, Combination of APACHE-II score and an obesity A resolution structure of the pancreatic lipase-colipase complex inhibited by a C11 alkyl
score (APACHE-O) for the prediction of severe acute pancreatitis. Pancreatology 4, 1–6 phosphonate. Biochemistry 34, 2751–2762 (1995).
(2004). 63. G. K. Veeramachaneni, K. K. Raj, L. M. Chalasani, J. S. Bondili, V. R. Talluri, High-throughput
39. G. I. Papachristou, D. J. Papachristou, H. Avula, A. Slivka, D. C. Whitcomb, Obesity virtual screening with e-pharmacophore and molecular simulations study
increases the severity of acute pancreatitis: Performance of APACHE-O score in the designing of pancreatic lipase inhibitors. Drug Des. Devel. Ther. 9, 4397–4412
and correlation with the inflammatory response. Pancreatology 6, 279–285 (2006). (2015).
40. B. De Waele, B. Vanmierlo, Y. Van Nieuwenhove, G. Delvaux, Impact of body overweight 64. S. M. Camhi, G. A. Bray, C. Bouchard, F. L. Greenway, W. D. Johnson, R. L. Newton,
and class I, II and III obesity on the outcome of acute biliary pancreatitis. Pancreas 32, E. Ravussin, D. H. Ryan, S. R. Smith, P. T. Katzmarzyk, The relationship of waist
343–345 (2006). circumference and BMI to visceral, subcutaneous, and total body fat: Sex and race
41. L. Sempere, J. Martinez, E. de Madaria, B. Lozano, J. Sanchez-Paya, R. Jover, M. Perez-Mateo, differences. Obesity (Silver Spring) 19, 402–408 (2011).
Obesity and fat distribution imply a greater systemic inflammatory response and a worse 65. J. Serth, A. Lautwein, M. Frech, A. Wittinghofer, A. Pingoud, The inhibition of the GTPase
prognosis in acute pancreatitis. Pancreatology 8, 257–264 (2008). activating protein-Ha-ras interaction by acidic lipids is due to physical association
42. R. Hegazi, A. Raina, T. Graham, S. Rolniak, P. Centa, H. Kandil, S. J. O’Keefe, Early jejunal of the C-terminal domain of the GTPase activating protein with micellar structures. EMBO
feeding initiation and clinical outcomes in patients with severe acute pancreatitis. JPEN J. 10, 1325–1330 (1991).
J. Parenter. Enteral Nutr. 35, 91–96 (2011). 66. P. Dumnicka, D. Maduzia, P. Ceranowicz, R. Olszanecki, R. Drożdż, B. Kuśnierz-Cabala, The
43. P. J. B. Davis, K. M. Eltawil, B. Abu-Wasel, M. J. Walsh, T. Topp, M. Molinari, Effect of obesity interplay between inflammation, coagulation and endothelial injury in the early phase

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


and decompressive laparotomy on mortality in acute pancreatitis requiring intensive of acute pancreatitis: Clinical implications. Int. J. Mol. Sci. 18, 354 (2017).
care unit admission. World J. Surg. 37, 318–332 (2013). 67. WHO Expert Consultation, Appropriate body-mass index for Asian populations and its
44. J. Katuchova, J. Bober, P. Harbulak, A. Hudak, T. Gajdzik, R. Kalanin, J. Radonak, Obesity implications for policy and intervention strategies. Lancet 363, 157–163 (2004).
as a risk factor for severe acute pancreatitis patients. Wien. Klin. Wochenschr. 126, 68. M. Deurenberg-Yap, G. Schmidt, W. A. van Staveren, P. Deurenberg, The paradox of low
223–227 (2014). body mass index and high body fat percentage among Chinese, Malays and Indians
45. E. Guzman Calderon, P. Montes Teves, E. Monge Salgado, Bisap-O: Obesity included in Singapore. Int. J. Obes. Relat. Metab. Disord. 24, 1011–1017 (2000).
in score BISAP to improve prediction of severity in acute pancreatitis. Rev. Gastroenterol. 69. E. J. Balthazar, D. L. Robinson, A. J. Megibow, J. H. Ranson, Acute pancreatitis: Value of CT
Peru 32, 251–256 (2012). in establishing prognosis. Radiology 174, 331–336 (1990).
46. J. Suazo-Baráhona, R. Carmona-Sánchez, G. Robles-Díaz, P. Milke-García, F. Vargas-Vorácková, 70. E. C. Rush, I. Freitas, L. D. Plank, Body size, body composition and fat distribution:
L. Uscanga-Domínguez, M. Peláez-Luna, Obesity: A risk factor for severe acute biliary Comparative analysis of European, Maori, Pacific Island and Asian Indian adults. Br.
and alcoholic pancreatitis. Am. J. Gastroenterol. 93, 1324–1328 (1998). J. Nutr. 102, 632–641 (2009).
47. Y. P. Yeung, B. Y. Lam, A. W. Yip, APACHE system is better than Ranson system 71. E. C. Rush, J. H. Goedecke, C. Jennings, L. Micklesfield, L. Dugas, E. V. Lambert, L. D. Plank,
in the prediction of severity of acute pancreatitis. Hepatobiliary Pancreat. Dis. Int. 5, BMI, fat and muscle differences in urban women of five ethnicities from two countries.
294–299 (2006). Int. J. Obes. (Lond) 31, 1232–1239 (2007).
48. Y. Yashima, H. Isayama, T. Tsujino, R. Nagano, K. Yamamoto, S. Mizuno, H. Yagioka, 72. J. van Grinsven, J. L. A. van Vugt, A. Gharbharan, T. L. Bollen, M. G. Besselink,
K. Kawakubo, T. Sasaki, H. Kogure, Y. Nakai, K. Hirano, N. Sasahira, M. Tada, T. Kawabe, H. C. van Santvoort, C. H. J. van Eijck, D. Boerma; Dutch Pancreatitis Study Group, The
K. Koike, M. Omata, A large volume of visceral adipose tissue leads to severe acute association of computed tomography-assessed body composition with mortality
pancreatitis. J. Gastroenterol. 46, 1213–1218 (2011). in patients with necrotizing pancreatitis. J. Gastrointest. Surg. 21, 1000–1008 (2017).
49. F. Yang, H. Wu, Y. Li, Z. Li, C. Wang, J. Yang, B. Hu, Z. Huang, R. Ji, X. Zhan, H. Xie, L. Wang, 73. P. Cohen, M. Miyazaki, N. D. Socci, A. Hagge-Greenberg, W. Liedtke, A. A. Soukas,
M. Zhang, C. Tang, Prevention of severe acute pancreatitis with octreotide in obese R. Sharma, L. C. Hudgins, J. M. Ntambi, J. M. Friedman, Role for stearoyl-CoA desaturase-1
patients: A prospective multi-center randomized controlled trial. Pancreas 41, 1206–1212 in leptin-mediated weight loss. Science 297, 240–243 (2002).
(2012). 74. R. J. Craven, R. W. Lencki, Crystallization, polymorphism, and binary phase behavior
50. S. R. Thomson, W. S. Hendry, G. A. McFarlane, A. I. Davidson, Epidemiology and outcome of model enantiopure and racemic triacylglycerols. Cryst. Growth Des. 11, 1723–1732
of acute pancreatitis. Br. J. Surg. 74, 398–401 (1987). (2011).
51. D. Yadav, A. B. Lowenfels, The epidemiology of pancreatitis and pancreatic cancer. 75. B. A. van Kuiken, W. D. Behnke, The activation of porcine pancreatic lipase by
Gastroenterology 144, 1252–1261 (2013). cis-unsaturated fatty acids. Biochim. Biophys. Acta 1214, 148–160 (1994).
52. T. L. Blasbalg, J. R. Hibbeln, C. E. Ramsden, S. F. Majchrzak, R. R. Rawlings, Changes 76. J. Folch, M. Lees, G. H. Sloane Stanley, A simple method for the isolation and purification
in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th of total lipides from animal tissues. J. Biol. Chem. 226, 497–509 (1957).
century. Am. J. Clin. Nutr. 93, 950–962 (2011). 77. W. R. Morrison, L. M. Smith, Preparation of fatty acid methyl esters and dimethylacetals
53. B. Khatua, R. N. Trivedi, P. Noel, K. Patel, R. Singh, C. de Oliveira, S. Trivedi, V. Mishra, from lipids with boron fluoride–methanol. J. Lipid Res. 5, 600–608 (1964).
M. Lowe, V. P. Singh, Carboxyl ester lipase may not mediate lipotoxic injury during severe 78. H. Ko, M. E. Royer, A gas-liquid chromatographic assay for plasma free fatty acids.
acute pancreatitis. Am. J. Pathol. 189, 1226–1240 (2019). J. Chromatogr. 88, 253–263 (1974).
54. C. de Oliveira, B. Khatua, A. Bag, B. El-Kurdi, K. Patel, V. Mishra, S. Navina, V. P. Singh, 79. L. S. Orlichenko, J. Behari, T.-H. Yeh, S. Liu, D. B. Stolz, A. K. Saluja, V. P. Singh,
Multimodal transgastric local pancreatic hypothermia reduces severity of acute Transcriptional regulation of CXC-ELR chemokines KC and MIP-2 in mouse pancreatic
pancreatitis in rats and increases survival. Gastroenterology 156, 735–747.e10 (2019). acini. Am. J. Physiol. Gastrointest. Liver Physiol. 299, G867–G876 (2010).
55. S. L. Blamey, C. W. Imrie, J. O'Neill, W. H. Gilmour, D. C. Carter, Prognostic factors in acute 80. H. M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T. N. Bhat, H. Weissig, I. N. Shindyalov,
pancreatitis. Gut 25, 1340–1346 (1984). P. E. Bourne, The Protein Data Bank. Nucleic Acids Res. 28, 235–242 (2000).
56. C. W. Imrie, I. S. Benjamin, J. C. Ferguson, A. J. McKay, I. Mackenzie, J. O'Neill,
81. S. Kim, J. Chen, T. Cheng, A. Gindulyte, J. He, S. He, Q. Li, B. A. Shoemaker, P. A. Thiessen,
L. H. Blumgart, A single-centre double-blind trial of trasylol therapy in primary acute
B. Yu, L. Zaslavsky, J. Zhang, E. E. Bolton, PubChem 2019 update: Improved access
pancreatitis. Br. J. Surg. 65, 337–341 (1978).
to chemical data. Nucleic Acids Res. 47, D1102–D1109 (2019).
57. M. Pini, J. A. Sennello, R. J. Cabay, G. Fantuzzi, Effect of diet-induced obesity on acute
82. E. Harder, W. Damm, J. Maple, C. Wu, M. Reboul, J. Y. Xiang, L. Wang, D. Lupyan,
pancreatitis induced by administration of interleukin-12 plus interleukin-18 in mice.
M. K. Dahlgren, J. L. Knight, J. W. Kaus, D. S. Cerutti, G. Krilov, W. L. Jorgensen, R. Abel,
Obesity (Silver Spring) 18, 476–481 (2010).
R. A. Friesner, OPLS3: A force field providing broad coverage of drug-like small molecules
58. J. A. Williams, M. Korc, R. L. Dormer, Action of secretagogues on a new preparation
and proteins. J. Chem. Theory Comput. 12, 281–296 (2016).
of functionally intact, isolated pancreatic acini. Am. J. Physiol. 235, 517–524 (1978).
59. E. Rogalska, S. Ransac, R. Verger, Stereoselectivity of lipases. II. Stereoselective hydrolysis
of triglycerides by gastric and pancreatic lipases. J. Biol. Chem. 265, 20271–20276 Acknowledgments: We thank J. N. Singh and R. Cannon for working with us on the
(1990). illustration shown in Fig. 7. Funding: This project was supported by R01DK092460 and
60. R. F. Oliveira, G. A. Gonçalves, F. D. Inácio, E. A. Koehnlein, C. G. M. De Souza, A. Bracht, R01DK119646 from the NIDDK and PR151612 and PR191945 from the DOD (to V.P.S.). Author
R. M. Peralta, Inhibition of pancreatic lipase and triacylglycerol intestinal absorption by contributions: V.P.S. designed, supervised, and conceptualized the study. Acquisition and

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 15 of 16


SCIENCE ADVANCES | RESEARCH ARTICLE

analysis of data were facilitated and carried out by B.K., B.E.-K., K.P., C.R., P.N., M.C., J.R.Y., S.K., Submitted 2 July 2020
A.G., M.L., and V.P.S.; while B.K., D.O.F., M.L., and V.P.S. critically evaluated the manuscript. Accepted 11 December 2020
Interpretation of data was done by B.K., B.E.-K., K.P., C.R., M.L., and V.P.S. Manuscript was Published 29 January 2021
drafted by B.K., B.E.-K., K.P., C.R., A.G., and V.P.S. Statistical analysis was done by B.K., B.E.-K., K.P., 10.1126/sciadv.abd6449
M.C., and V.P.S. Funding was obtained by V.P.S. Competing interests: The authors declare
that they have no competing interests. Data and materials availability: All data needed to Citation: B. Khatua, B. El-Kurdi, K. Patel, C. Rood, P. Noel, M. Crowell, J. R. Yaron, S. Kostenko, A. Guerra,
evaluate the conclusions in the paper are present in the paper and/or the Supplementary D. O. Faigel, M. Lowe, V. P. Singh, Adipose saturation reduces lipotoxic systemic inflammation
Materials. Additional data related to this paper may be requested from the authors. and explains the obesity paradox. Sci. Adv. 7, eabd6449 (2021).

Downloaded from http://advances.sciencemag.org/ on February 2, 2021

Khatua et al., Sci. Adv. 2021; 7 : eabd6449 29 January 2021 16 of 16


Adipose saturation reduces lipotoxic systemic inflammation and explains the obesity paradox
Biswajit Khatua, Bara El-Kurdi, Krutika Patel, Christopher Rood, Pawan Noel, Michael Crowell, Jordan R. Yaron, Sergiy
Kostenko, Andre Guerra, Douglas O. Faigel, Mark Lowe and Vijay P. Singh

Sci Adv 7 (5), eabd6449.


DOI: 10.1126/sciadv.abd6449

Downloaded from http://advances.sciencemag.org/ on February 2, 2021


ARTICLE TOOLS http://advances.sciencemag.org/content/7/5/eabd6449

SUPPLEMENTARY http://advances.sciencemag.org/content/suppl/2021/01/25/7.5.eabd6449.DC1
MATERIALS

REFERENCES This article cites 81 articles, 9 of which you can access for free
http://advances.sciencemag.org/content/7/5/eabd6449#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science Advances (ISSN 2375-2548) is published by the American Association for the Advancement of Science, 1200 New
York Avenue NW, Washington, DC 20005. The title Science Advances is a registered trademark of AAAS.
Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of
Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial
License 4.0 (CC BY-NC).

You might also like