ArticlePDF Available

Sex differences in islet stress responses support female β cell resilience

Authors:

Abstract and Figures

Objective: Pancreatic β cells play a key role in maintaining glucose homeostasis; dysfunction of this critical cell type causes type 2 diabetes (T2D). Emerging evidence points to sex differences in β cells, but few studies have examined male-female differences in β cell stress responses and resilience across multiple contexts, including diabetes. Here, we address the need for high-quality information on sex differences in β cell and islet gene expression and function using both human and rodent samples. Methods: We compared β cell gene expression and insulin secretion in donors with T2D to non-diabetic donors in both males and females. In mice, we generated a well-powered islet RNAseq dataset from 20-week-old male and female siblings with similar insulin sensitivity. Our unbiased analysis of murine gene expression pointed to a sex difference in the endoplasmic reticulum (ER) stress response. Based on this analysis, we hypothesize female islets will be more resilient to ER stress than male islets. To test this, we subjected islets isolated from age-matched male and female mice to thapsigargin treatment and monitored protein synthesis, cell death, and β cell insulin production and secretion. Transcriptomic and proteomic analyses were used to characterize sex differences in islet responses to ER stress. Results: Our single-cell analysis of human β cells revealed sex-specific changes to gene expression and function in T2D, correlating with more robust insulin secretion in human islets isolated from female donors with T2D compared to male donors with T2D. In mice, RNA sequencing revealed differential enrichment of unfolded protein response pathway-associated genes, where female islets showed higher expression of genes linked with protein synthesis, folding, and processing. This differential expression was biologically significant, as islets isolated from female mice were more resilient to ER stress induction with thapsigargin. Specifically, female islets showed a greater ability to maintain glucose-stimulated insulin production and secretion during ER stress compared with males. Conclusions: Our data demonstrate sex differences in β cell gene expression in both humans and mice, and that female β cells show a greater ability to maintain glucose-stimulated insulin secretion across multiple physiological and pathological contexts.
Sex differences in human islet transcriptomic and functional responses in type 2 diabetes. scRNAseq data from male and female human b cells. For donor metadata see Supplementary file 8. (AeC) Venn diagrams compare the number of significantly differentially expressed genes between ND and T2D donors (p-adj<0.05). All differentially expressed genes (A), downregulated genes (B), upregulated genes (C) in T2D human b cells. For complete gene lists see Supplementary file 1 and 2. (DeF) Top 10 significantly enriched Reactome pathways (ND vs T2D) from non-sex-specific (D), female (E), or male (F) significantly differentially expressed genes (p-adj< 0.05). Gene ratio is calculated as k/n, where k is the number of genes identified in each Reactome pathway, and n is the number of genes from the submitted gene list participating in any Reactome pathway. For complete Reactome pathway lists see Supplementary file 2. (GeK) Human islet perifusion data from the Human Pancreas Analysis Program in ND and T2D donor islets in females (F, I) and males (G, H). 3 mM glucose (3 mM G); 16.7 mM glucose (16.7 mM G); 0.1 mM isobutylmethylxanthine (0.1 mM IBMX); 30 mM potassium chloride (30 mM KCl); 4 mM amino acid mixture (4 mM AAM; mM: 0.44 alanine, 0.19 arginine, 0.038 aspartate, 0.094 citrulline, 0.12 glutamate, 0.30 glycine, 0.077 histidine, 0.094 isoleucine, 0.16 leucine, 0.37 lysine, 0.05 methionine, 0.70 ornithine, 0.08 phenylalanine, 0.35 proline, 0.57 serine, 0.27 threonine, 0.073 tryptophan, and 0.20 valine, 2 mM glutamine). (IeK) Quantification of area under the curve (AUC) is shown for the various stimulatory media in females (I), males (J) and donors with T2D (K). (I) In females, insulin secretion from ND islets was not significantly higher than T2D islets under any culture condition (p ¼ 0.4806 [AAM þ LG], p ¼ 0.2270 [AAM þ HG], p ¼ 0.1384 [AAM þ HG þ IBMX], and p ¼ 0.1465 [KCl]; unpaired Student's t-test). (J) In males, insulin secretion from ND islets was significantly higher than T2D islets under 4 mM AAM þ16.7 mM glucose (HG) þ 0.1 mM IBMX stimulation (p ¼ 0.0442 [AAM þ HG þ IBMX]; unpaired Student's t-test), but not in other conditions (p ¼ 0.5315 [AAM þ LG], p ¼ 0.0818 [AAM þ HG], and p ¼ 0.2259 [KCl]; unpaired Student's t-test). (K) Total insulin secretion showed a trend toward lower secretion in T2D male islets than ND male islets (p ¼ 0.1514 and p ¼ 0.0503 for females and males, respectively; unpaired Student's t-test). * indicates p < 0.05; ns indicates not significant; error bars indicate SEM.
… 
Sex differences in ex vivo and in vivo insulin secretion. (A) Experimental workflow of static glucose-stimulated insulin secretion. (B, C) Relative high glucose (20 mM; high glucose, HG) in treatments compared with DMSO in female (B) and male (C) islets. Female islet HG secretion was significantly higher compared with control after 0-and 2-hour Tg pre-treatments (p ¼ 0.0083 [0-hour] and p ¼ 0.0371 [2-hour]; Mann Whitney test). Male islet HG secretion was significantly lower compared with control after a 4-hour Tg pre-treatment (p ¼ 0.0013; Mann Whitney test). (D) Insulin content. Female islet insulin content was significantly higher compared with control after a 4-hour Tg pre-treatment (p ¼ 0.0269; Mann Whitney test). (E) Proinsulin content. Female islet proinsulin content was significantly lower compared with control after a 2-hour Tg pre-treatment (p ¼ 0.0437; Mann Whitney test). Male islet proinsulin content was significantly lower compared with control after 2-and 4-hour Tg pre-treatments (p ¼ 0.0014 [2-hour] and p ¼ 0.0005 [4-hour]; Mann Whitney test). (FeH) Physiology measurements after a 6-hour fast in 20-week-old male and female B6 mice. (F, G) Insulin levels from glucose-stimulated insulin secretion tests (F: nM, G: % basal insulin) following a single glucose injection (2 g glucose/kg body weight, i.p). Area under the curve (AUC) calculations (n ¼ 13 females, n ¼ 18 males). (F) Insulin levels were significantly higher in male mice at 0 min and 30 min post injection (p ¼ 0.0063 [0 min] and p ¼ 0.0009 [30 min]; Student's t-test). AUC was significantly higher in males (p ¼ 0.0159; Student's t-test). (G) Insulin levels (% baseline). Glucose-stimulated insulin secretion was significantly higher in female mice 15 min post injection (p ¼ 0.0279; Student's t-test). (H) Glucose levels from glucose tolerance tests following a single glucose injection (2 g glucose/kg body weight). AUC calculations (n ¼ 11 females, n ¼ 11 males). For B-E, grey triangles indicate the concentration of insulin or proinsulin from five islets, black circles indicate the average values per mouse. * indicates p < 0.05, ** indicates p < 0.01, *** indicates p < 0.001; ns indicates not significant; error bars indicate SEM.
… 
Content may be subject to copyright.
Sex differences in islet stress responses support
female
b
cell resilience
George P. Brownrigg, Yi Han Xia, Chieh Min Jamie Chu, Su Wang, Charlotte Chao, Jiashuo Aaron Zhang,
Søs Skovsø, Evgeniy Panzhinskiy, Xiaoke Hu, James D. Johnson
*
, Elizabeth J. Rideout
*
ABSTRACT
Objective: Pancreatic
b
cells play a key role in maintaining glucose homeostasis; dysfunction of this critical cell type causes type 2 diabetes
(T2D). Emerging evidence points to sex differences in
b
cells, but few studies have examined male-female differences in
b
cell stress responses
and resilience across multiple contexts, including diabetes. Here, we address the need for high-quality information on sex differences in
b
cell and
islet gene expression and function using both human and rodent samples.
Methods: In humans, we compared
b
cell gene expression and insulin secretion in donors with T2D to non-diabetic donors in both males and
females. In mice, we generated a well-powered islet RNAseq dataset from 20-week-old male and female siblings with similar insulin sensitivity.
Our unbiased gene expression analysis pointed to a sex difference in the endoplasmic reticulum (ER) stress response. Based on this analysis, we
hypothesized female islets would be more resilient to ER stress than male islets. To test this, we subjected islets isolated from age-matched male
and female mice to thapsigargin treatment and monitored protein synthesis, cell death, and
b
cell insulin production and secretion. Tran-
scriptomic and proteomic analyses were used to characterize sex differences in islet responses to ER stress.
Results: Our single-cell analysis of human
b
cells revealed sex-specic changes to gene expression and function in T2D, correlating with more
robust insulin secretion in human islets isolated from female donors with T2D compared to male donors with T2D. In mice, RNA sequencing
revealed differential enrichment of unfolded protein response pathway-associated genes, where female islets showed higher expression of genes
linked with protein synthesis, folding, and processing. This differential expression was physiologically signicant, as islets isolated from female
mice were more resilient to ER stress induction with thapsigargin. Specically, female islets showed a greater ability to maintain glucose-
stimulated insulin production and secretion during ER stress compared with males.
Conclusions: Our data demonstrate sex differences in
b
cell gene expression in both humans and mice, and that female
b
cells show a greater
ability to maintain glucose-stimulated insulin secretion across multiple physiological and pathological contexts.
Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Keywords Pancreatic islets;
b
cells; Diabetes mellitus; Endoplasmic reticulum stress; Protein synthesis; Transcriptomics
1. INTRODUCTION
Pancreatic
b
cells make and secrete insulin, an essential hormone
required to maintain whole-body glucose homeostasis. Emerging evi-
dence from multiple species points to biological sex as an important, but
often overlooked, factor that affects
b
cell biology [1e6]. Large-scale
surveys of gene expression in mice and humans show that differ-
ences exist between the sexes in the pancreas [7e9], in islets [10], and
in
b
cells specically [4,11]. Humans also have a sex-specic
b
cell
gene expression response to aging [12], and show male-female dif-
ferences in pancreatic
b
cell number [6]. With respect to
b
cell function,
most data from rodent and human studies suggests glucose-stimulated
insulin secretion is higher in females than in males [5,10,13e16]. While
male-female differences in peripheral insulin sensitivity [15,17e27]may
contribute to these differences, sex-biased insulin secretion in humans
persists in the context of equivalent insulin sensitivity between males
and females [5]. Whether sex differences in other aspects of
b
cell gene
expression and function are similarly independentof insulin sensitivity in
rodents and humans remains unclear, as insulin sensitivity is not
routinely monitored across datasets showing sex differences in
b
cell
biology.
Biological sex also affects the risk of developing T2D. Across many
population groups, men are at a higher risk of developing T2D than
women [28e31]. Some of this differential risk is explained by lifestyle
and cultural factors [31e33]. Biological sex also plays a role; however,
as the male-biased risk of developing diabetes-like phenotypes is
conserved across multiple animal models [22,34e39]. Despite a
dominant role for
b
cell function in T2D pathogenesis [40,41], T2D-
and stress-associated changes to
b
cell gene expression and function
in each sex remain largely unexplored, as most studies on these topics
do not include biological sex as a variable in their analysis [42e49].
Collecting detailed knowledge of
b
cell gene expression and function
from each sex under physiological and pathological conditions is
therefore a key rst step toward understanding whether male-female
differences in this important cell type may contribute to T2D risk.
The overall goal of our study was to provide detailed knowledge of
b
cell gene expression and function in both males and females across
multiple contexts to advance our understanding of sex differences in
Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
*Corresponding authors. E-mails: james.d.johnson@ubc.ca (J.D. Johnson), elizabeth.rideout@ubc.ca (E.J. Rideout).
Received October 12, 2022
Revision received January 7, 2023
Accepted January 17, 2023
Available online 20 January 2023
https://doi.org/10.1016/j.molmet.2023.101678
Original Article
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 1
this important cell type. Our data show male-female differences in
islet and
b
cell gene expression and stress responses in both
humans and mice. These differences contribute to sex differences in
b
cell resilience, where we nd female
b
cells show a greater ability
to maintain glucose-stimulated insulin secretion in response to stress
and T2D in mice and humans, respectively. Given that an insulin
tolerance test indicated similar insulin sensitivity between the male
and female mice used in our study, our ndings suggest biological
sex is an important variable to consider in studies on islet and
b
cell
function.
2. MATERIALS AND METHODS
2.1. Animals
Mice were bred in-house or purchased from the Jackson Laboratory.
Unless otherwise stated, islets were isolated from C57BL/6J mice aged
20e24 weeks. Animals were housed and studied in the UBC Modied
Barrier Facility using protocols approved by the UBC Animal Care
Committee and in accordance with international guidelines. Mice were
housed on a 12-hour light/dark cycle with food and drinking water ad
libitum. Mice were fed a regular chow diet (LabDiet #5053); 24.5%
energy from protein, 13.1% energy from fat, and 62.4% energy from
carbohydrates.
2.2. Islet isolation, culture, dispersion and treatment
Mouse islet isolations were performed by ductal collagenase injection
followed by ltration and hand-picking, using modications of the
protocol described by Salvalaggio [50]. Islets recovered overnight, in
islet culture media (RPMI media with 11.1 mM
D
-glucose supple-
mented with 10% vol/vol fetal bovine serum (FBS) (Thermo:
12483020) and 1% vol/vol Penicillin-Streptomycin (P/S) (GIBCO:
15140-148)) at 37
C with 5% CO
2
. After four washes with Minimal
Essential Medium [
L
-glutamine, calcium and magnesium free] (Corn-
ing: 15-015 CV) islets were dispersed with 0.01% trypsin and
resuspended in islet culture media. Cell seedings were done as per the
experimental procedure (protein synthesis: 20,000 cells per well, live
cell imaging: 5,000 cells per well). ER stress was induced by treating
islets with the SERCA inhibitor thapsigargin. For assays less than 24 h,
we used (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S). For assays greater
than 24 h we used (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S, 10% vol/
vol FBS).
2.3. Analysis of protein synthesis
Dispersed islets were seeded into an optical 96-well plate (Perkin
Elmer) at a density of approximately 20,000 cells per well in islet
culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S, 10% vol/
vol FBS). 24 h after seeding, treatments were applied in fresh islet
culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S). After in-
cubation, fresh culture media was applied (11.1 mM
D
-glucose
RPMI, 1% vol/vol P/S), then supplemented with 20
m
MOPP(Invi-
trogen) and respective drug treatments. The assay was performed
according to manufacturers instructions. Cells were imaged at 10
with an ImageXpress
MICRO
high-content imager and analyzed with
MetaXpress (Molecular Devices) to quantify the integrated staining
intensity of OPP-Alexa Fluor 594 in cells identied by NuclearMask
Blue Stain.
2.4. Live cell imaging
Dispersed islets were seeded into 384-well plates (Perkin Elmer) at a
density of approximately 5,000 cells per well. Islet viability was
measured with the TC20 Automated Cell Counter (Bio-Rad: 1450102)
with Trypan Blue (Bio-Rad: 1450021). Islets were allowed to adhere for
48 h in islet culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S,
10% vol/vol FBS). Cells were stained with Hoechst 33342 (Sigmae
Aldrich) (0.05
m
g/mL) and propidium iodide (SigmaeAldrich) (0.5
m
g/
mL) for 1 h in islet culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol
P/S, 10% vol/vol FBS) prior to the addition of treatments and imaging.
384-well plates were placed into the environmentally-controlled
(37
C, 5% CO2) ImageXpress
MICRO
high content imaging system. To
measure cell death, islet cells were imaged every 2 h for 84 h. Met-
aXpress software was used to quantify cell death, dened as the
number of Propidium Iodide-positive/Hoechst 33342-positive cells. To
measure Ins2 gene activity, dispersed islets from Ins2
GFP/WT
mice aged
21e23 weeks were used [51]. Islet cells were imaged every 30 min for
60 h. MetaXpress analysis software and custom R scripts were used to
perform single-cell tracking of Ins2
GFP/WT
b
cells as previously
described [51].
2.5. Western blot
After overnight recovery, islets were split equally per mouse into islet
culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S, 10% vol/vol
FBS) resulting in w100e150 islets per condition. Islets were treated
for 24 h with DMSO or 1
m
M Tg in islet culture media then sonicated in
RIPA lysis buffer (150 mM NaCl, 1% Nonidet P-40, 0.5% DOC, 0.1%
SDS, 50 mM Tris (pH 7.4), 2 mM EGTA, 2 mM Na
3
VO
4
, and 2 mM NaF
supplemented with complete mini protease inhibitor cocktail (Roche,
Laval, QC)). Equal protein amounts (8e10
m
g of protein) in equal
volumes were loaded for each experiment. Protein lysates were
incubated in Laemmli loading buffer (Thermo, J61337AC) at 95
C for
5 min and resolved by SDS-PAGE. Proteins were then transferred to
PVDF membranes (BioRad, CA) and probed with antibodies against
HSPA5 (1:1000, Cat. #3183, Cell Signalling), eIF2
a
(1:1000, Cat.
#2103, Cell Signalling), phospho-eIF2
a
(1:1000, Cat. #3398, Cell
Signalling), IRE1
a
(1:1000, Cat. #3294, Cell Signalling), phospho-
IRE1
a
(1:1000, Cat. #PA1-16927, Thermo Fisher Scientic), CHOP
(1:1000, #ab11419, Abcam),
b
-actin (1:1000, NB600-501, Novus
Biologicals). The signals were detected by secondary HRP-conjugated
antibodies (Anti-mouse, Cat. #7076; Anti-rabbit, Cat. #7074; CST) and
either Pierce ECL Western Blotting Substrate (Thermo Fisher Scientic)
or Forte (Immobilon). Protein band intensities were quantied using
Image Studio (LI-COR).
2.6. Islet secretion and content
Glucose-stimulated insulin/proinsulin production and secretion were
assessed using size-matched islets (ve islets per well, in triplicate)
seeded into 96-well V-bottom Tissue Culture Treated Microplates
(Corning: #CLS3894). Islets were allowed to adhere for 48 h in culture
media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S, 10% vol/vol FBS),
based on a published method [52], as this protocol permits analysis of
large sample numbers and treatments, and minimizes islet loss.
Adherent islets were washed with KrebseRinger Buffer (KRB; 129 mM
NaCl, 4.8 mM KCl, 1.2 mM MgSO
4
, 1.2 mM KH
2
PO
4
, 2.5 mM CaCl
2
,
5 mM NaHCO
3
, 10 mM HEPES, 0.5% bovine serum albumin) con-
taining 3 mM glucose then pre-incubated for 4 h in 3 mM glucose KRB.
1
m
M Tg was added to the 3 mM low glucose pre-incubation buffer 4 h
prior, 2 h prior, or at the start of the low glucose incubation period.
Islets were incubated in KRB with 3 mM glucose then 20 mM glucose
for 45 min each. Supernatant was collected after each stimulation. Islet
insulin and proinsulin content was extracted by freeze-thawing in
100
m
L of acid ethanol, then the plates were shaken at 1200 rpm for
10 min at 4
C to lyse the islets. Insulin was measured by Rodent
Insulin Chemiluminescent ELISA (ALPCO: 80-INSMR) and proinsulin by
Original Article
2MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
Rat/Mouse Proinsulin ELISA (Mercodia: 10-1232-01). Measurements
were performed on a Spark plate reader (TECAN).
2.7. Blood collection and in vivo analysis of glucose homeostasis
and insulin secretion
Mice were fasted for 6 h prior to glucose and insulin tolerance tests.
During glucose and insulin tolerance tests, tail blood was collected
for blood glucose measurements using a glucometer (One Touch
Ultra 2 Glucometer, Lifescan, Canada). For intraperitoneal (i.p.)
glucose tolerance tests, the glucose dose was 2 g glucose/kg of body
mass. For insulin tolerance tests, the insulin dose was 0.75U insulin/
kg body mass. For measurements of in vivo glucose-stimulated in-
sulin secretion, femoral blood was collected after i.p. injection of 2 g
glucose/kg body mass. Blood samples were kept on ice during
collection, centrifuged at 2000 rpm for 10 min at 4
C and stored as
plasma at 20
C. Plasma samples were analysed for insulin using
Rodent Insulin Chemiluminescent ELISA (ALPCO: 80-INSMR).
Glucose-stimulated insulin secretion from an independent large
cohort of Insr
f/f
:Ins1Cre
/
:nTnG
þ/
mice reared in our facility was
replotted from a published Johnson lab study [53]. Blood glucose
was monitored using control and insulin-reduced mice. Control mice
were Ins1
/
:Ins2
f/f
:mTmG þtamoxifen and Ins1
/
:Ins2
f/f
:
Pdx1Cre
ERT
:mTmG þcorn oil [54]. Because we conrmed there were
no signicant differences in blood glucose between control geno-
types, the data were combined in our analysis. Insulin-reduced mice
were generated by injecting Ins1
/
:Ins2
f/f
:Pdx1Cre
ERT
:mTmG mice
with tamoxifen (3 mg/40 g body weight, dissolved in corn oil, for 4
consecutive days) at 6e8 weeks [54].
2.8. RNA sequencing
To assess basal transcriptional differences, islets from male and fe-
male mice (n ¼9M, 8F) were snap-frozen and stored at 80
C until
RNA extraction. To assess Tg-induced transcriptional changes, islets
from each mouse were treated with DMSO or 1
m
M Tg for 6- or 12-
hours in culture media (11.1 mM
D
-glucose RPMI, 1% vol/vol P/S).
Four groups per sex (eight groups total) were analyzed: 6 h DMSO,
6 h Tg, 12 h DMSO and 12 h Tg, n ¼3e4 per group, each nrep-
resents pooled islet RNA from two mice. Islets were frozen at 80
Cin
100
m
L of RLT buffer (Qiagen) with beta mercaptoethanol (1%). RNA
was isolated using RNeasy Mini Kit (Qiagen #74106) according to
manufacturers instructions. RNA from 43 to 62 islets was pooled from
two mice and 19e150 ng of RNA was sequenced per pooled sample.
RNA sequencing was performed at the UBC Biomedical Research
Centre Sequencing Core. Sample quality control was performed using
the Agilent 2100 Bioanalyzer System (RNA Pico LabChip Kit). Qualifying
samples were prepped following the standard protocol for the NEBNext
Ultra II Stranded mRNA (New England Biolabs). Sequencing was per-
formed on the Illumina NextSeq 500 with Paired End 42bp 42bp
reads. Demultiplexed read sequences were then aligned to the
reference sequence (UCSC mm10) using STAR aligner (v 2.5.0b) [55].
Gene differential expression was analyzed using DESeq2 R package
[56]. Pathway enrichment analysis were performed using Reactome
[57]. Over-representation analysis was performed using NetworkA-
nalyst3.0 (www.networkanalyst.ca)[58].
2.9. Proteomics
Islets were treated with DMSO or 1
m
M Tg for 6 h in islet culture media
(11.1 mM
D
-glucose RPMI, 1% vol/vol P/S). Two groups per sex (four
groups total): 6 h DMSO and 6 h Tg, n ¼5e7 per group, each n
represents 200e240 islets pooled from two mice. Islet pellets were
frozen at 80
C in 100
m
L of SDS lysis buffer (4% SDS, 100 mM Tris,
pH 8) and the proteins in each sample were precipitated using acetone.
The University of Victoria proteomics service performed non-targeted
quantitative proteomic analysis using data-independent acquisition
(DIA) with LC-MS/MS on an Orbitrap mass spectrometer using 1
m
gof
protein. A mouse FASTA database was downloaded from Uniprot
(http://uniprot.org). This le was used with the 6 gas phase fraction
les from the analysis of the chromatogram library sample to create a
mouse islet-specic chromatogram library using the EncyclopeDIA (v
1.2.2) software package (Searle et al., 2018). This chromatogram li-
brary le was then used to perform identication and quantitation of
the proteins in the samples again using EncyclopeDIA with Overlapping
DIA as the acquisition type, trypsin used as the enzyme, CID/HCD as
the fragmentation, 10 ppm mass tolerances for the precursor, frag-
ment, and library mass tolerances. The Percolator version used was
3.10. The precursor FDR rate was set to 1%. Protein abundances were
log2 transformed, imputation was performed for missing values, then
proteins were normalized to median sample intensities. Differential
expression was analyzed using limma in Perseus [59].
2.10. Analysis of the transcriptome and partial proteome
Tg-induced changes to gene expression and protein levels were
compared 6 h post-treatment. Log2-transformed fold change values
were used to assess the congruence between our proteomics data and
RNAseq data. Genes and proteins that were concordantly altered by Tg
treatment at both the mRNA and protein level were searched in
PubMed for relevant literature on their role in
b
cells. The search term
used was ((beta cell) OR (islet) OR (
b
cell)) AND (Gene_Name).
Additional annotations for all mouse proteins were downloaded from
Uniprot [Dec 2022].
2.11. Data from HPAP
To compare sex differences in dynamic insulin secretion, data acquired
was from the Human Pancreas Analysis Program (HPAP-
RRID:SCR_016202) Database (https://hpap.pmacs.upenn.edu), a Hu-
man Islet Research Network (RRID:SCR_014393) consortium (UC4-
DK-112217, U01-DK-123594, UC4-DK-112232, and U01-DK-
123716).
2.12. Statistical analysis
Statistical analyses and data presentation were carried out using
GraphPad Prism 9 (GraphPad Software, San Diego, CA, USA) or R (v
4.1.1). Correlation plots were generated using the corrplot R package
(v 0.92) with default settings [60]. All R codes are published on github
(https://github.com/johnsonlabubc/ER-Stress-in-Mouse-Beta-Cells-
Data-Analysis). Studentst-tests or two-way ANOVAs were used for
parametric data. A ManneWhitney test was used for non-parametric
data. Statistical tests are indicated in the gure legends. For all sta-
tistical analyses, differences were considered signicant if the p-value
was less than 0.05. *:p<0.05; **p<0.01; ***p<0.001. Data
were presented as means SEM with individual data points from
biological replicates.
3. RESULTS
3.1. Sex differences in
b
cell transcriptional and functional
responses in ND and T2D human islets
Gene expression studies on human pancreas and islets identify sig-
nicant sex differences in gene expression [7,10]. Indeed, >1500
genes expressed in the pancreas show sex-biased expression [7].
While our dataset contained fewer individuals, we also note sex dif-
ferences in mRNA levels of many genes in
b
cells (Supplementary le
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 3
1). Given this differential gene expression, we wanted to dene
b
cell-
specic gene expression changes in T2D in each sex. We therefore
used a recently-compiled meta-analysis of publicly available scRNAseq
datasets from male and female human islets [61]. Our goal was to use
sex-based analysis to determine whether
b
cell gene expression
changes in T2D are shared between the sexes. In line with prior reports
[12],
b
cells from non-diabetic (ND) and T2D donors showed signi-
cant transcriptional differences. In
b
cells isolated from female T2D
donors, mRNA levels of 127 genes were signicantly different from ND
female donors (77 downregulated, 50 upregulated in T2D) (Figure 1Ae
C). In
b
cells isolated from male T2D donors, 462 genes were
differentially expressed compared with male ND donors (138 down-
regulated, 324 upregulated in T2D) (Figure 1AeC). Of the 660 genes
that were differentially regulated in T2D, 71 were differentially regu-
lated in both males and females (15 downregulated, 56 upregulated in
T2D) (Figs. S1AeC); however, the fold change for these 71 shared
genes was different between males and females (Fig. S1A; Supple-
mentary le 2). This suggests that for shared genes, the magnitude of
gene expression changes in T2D was not the same between the sexes.
Beyond shared genes, we observed that the majority of differentially
expressed genes in T2D (589/660) were unique to either males or
females (Figs. S1B,C; Supplementary le 2). Indeed, the most prom-
inent gene expression changes in T2D were found in genes that were
unique to one sex (Figs. S2A,B; Supplementary le 2). While these data
do not address the reasons for the sex-biased risk of T2D, and could
reect differences in medication [62e65], age [12,16,66e68], and
body mass index [15,68], our data suggest biological sex inuences
b
cell gene expression in T2D.
To determine which biological pathways were altered in
b
cells of T2D
donors from each sex, we performed pathway enrichment analysis.
Genes that were upregulated in
b
cells isolated from T2D donors
included genes involved in Golgi-ER transport and the unfolded protein
response (UPR) pathways (Figure 1DeF; Supplementary le 2). While
these biological pathways were signicantly upregulated in T2D in both
males and females, w75% of the differentially regulated genes in
these categories were unique to each sex (Table 1). Genes that were
downregulated in
b
cells from T2D donors revealed further differences
between the sexes: biological pathways downregulated in
b
cells from
female T2D donors included cellular responses to stress and to stimuli
(Figure 1E; Supplementary le 2), whereas
b
cells from male T2D
donors showed downregulation of pathways associated with respira-
tory electron transport and translation initiation (Figure 1F; Supple-
mentary le 2). Our analysis therefore suggests that sex-biased
b
cell
gene expression responses to T2D may inuence different cellular
processes in males and females.
The sex-biased
b
cell transcriptional response in T2D prompted us to
compare glucose-stimulated insulin secretion in each sex from ND and
T2D human islets using data from the Human Pancreas Analysis
Program database [69]. In ND donors, islets from males and females
showed similar patterns of insulin secretion in response to various
stimulatory media (Figure 1G,H). In donors with T2D, we found that
insulin secretion was impaired to a greater degree in islets from males
than in females (Figure 1GeK). This difference cannot be fully attrib-
uted to a sex difference in disease severity, as our analysis of donor
characteristics revealed no signicant correlation between sex and
HbA1c (Fig. S3A). Indeed, in male but not female islets, insulin
secretion was lower in donors with T2D following stimulation with both
high glucose and IBMX (Figure 1I,J), which potentiates insulin secre-
tion by increasing cAMP levels to a similar degree as the incretins [70].
Human islets from female donors with T2D therefore show a greater
ability to maintain glucose-stimulated insulin secretion than islets from
males with T2D (Figure 1K). Indeed, while diabetes status was the
main donor characteristic that correlated with changes in insulin
secretion (Fig. S3B), we noted that in T2D sex and age were two donor
characteristics showing trends toward an effect on insulin secretion
(Fig. S3A). Combined with our
b
cell gene expression data, these
ndings suggest that
b
cell transcriptional and functional responses in
T2D are not shared between the sexes.
3.2. Sex differences in UPR-associated gene expression in mouse
islets
Our unbiased analysis of human
b
cell gene expression and function in
T2D revealed differences between male and female donors with T2D.
Because human
b
cell gene expression and function can be affected by
factors such as peripheral insulin sensitivity, disease processes, and
medication [31,33], we investigated sex differences in
b
cell gene
expression and function in another context. We generated a well-
powered islet RNAseq dataset from 20-week-old male and female
C57BL/6J mice. We used an insulin tolerance test (ITT) to show that
insulin sensitivity was similar between the sexes at this age (Fig. S4A);
however, we acknowledge that the ITT may not be as sensitive as a
hyperinsulinemic-euglycemic clamp in detecting modest sex differ-
ences in insulin sensitivity.
Principal component analysis and unsupervised clustering clearly
separated male and female islets on the basis of gene expression
(Figure 2A; Fig. S5A). We found that 17.7% (3268/18938) of genes
were differentially expressed between the sexes (1648 upregulated in
females, 1620 upregulated in males), in line with estimates of sex-
biased gene expression in other tissues [71,72]. Overrepresentation
and pathway enrichment analysis both identied UPR-associated
pathways as a biological process that differed signicantly between
the sexes, where the majority of genes in this category were enriched
in female islets (Figure 2B,C; Supplementary le 3). Additional genes
that were enriched in female islets were those associated with the
gene ontology term Cellular response to endoplasmic reticulum
stress(GO:0034976), which included many genes involved in regu-
lating protein synthesis (Figure 2D). For example, females showed
signicantly higher levels of most ribosomal protein genes (Figure 2E).
Further genes enriched in females included those associated with
protein folding, protein processing, and quality control (Figure 2D).
Given that protein synthesis, processing, and folding capacity are
intrinsically important for multiple islet cell types [73e76], including
b
cells [77,78], this suggests female islets may have a larger protein
production and folding capacity than male islets.
3.3. Female islets are more resilient to endoplasmic reticulum
stress in mice
The burden of insulin production causes endoplasmic reticulum (ER)
stress in
b
cells [79e81]. ER stress is associated with an attenuation
of mRNA translation [82], and, if ER stress is prolonged, can lead to cell
death [83e85]. Given that female islets exhibited higher expression of
genes associated with protein synthesis, processing, and folding than
males, and higher expression of genes associated with the UPR, which
is activated in response to ER stress [86], we examined global protein
synthesis rates in male and female islets under basal conditions and
during ER stress. We incubated islets with O-propargyl-puromycin
(OPP), which is incorporated into newly-translated proteins and can be
ligated to a uorophore. Using this technique, we monitored the
accumulation of newly-synthesized islet proteins with single-cell
resolution (Fig. S6A). In basal culture conditions, male islet cells had
signicantly greater protein synthesis rates compared with female islet
cells (Fig. S6B). To investigate islet protein synthesis under ER stress in
Original Article
4MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
Figure 1: Sex differences in human islet transcriptomic and functional responses in type 2 diabetes. scRNAseq data from male and female human
b
cells. For donor
metadata see Supplementary le 8. (AeC) Venn diagrams compare the number of signicantly differentially expressed genes between ND and T2D donors (p-adj<0.05). All
differentially expressed genes (A), downregulated genes (B), upregulated genes (C) in T2D human
b
cells. For complete gene lists see Supplementary le 1 and 2. (DeF) Top 10
signicantly enriched Reactome pathways (ND vs T2D) from non-sex-specic (D), female (E), or male (F) signicantly differentially expressed genes (p-adj<0.05). Gene ratio is
calculated as k/n, where kis the number of genes identied in each Reactome pathway, and nis the number of genes from the submitted gene list participating in any Reactome
pathway. For complete Reactome pathway lists see Supplementary le 2. (GeK) Human islet perifusion data from the Human Pancreas Analysis Program in ND and T2D donor
islets in females (F, I) and males (G, H). 3 mM glucose (3 mM G); 16.7 mM glucose (16.7 mM G); 0.1 mM isobutylmethylxanthine (0.1 mM IBMX); 30 mM potassium chloride
(30 mM KCl); 4 mM amino acid mixture (4 mM AAM; mM: 0.44 alanine, 0.19 arginine, 0.038 aspartate, 0.094 citrulline, 0.12 glutamate, 0.30 glycine, 0.077 histidine, 0.094
isoleucine, 0.16 leucine, 0.37 lysine, 0.05 methionine, 0.70 ornithine, 0.08 phenylalanine, 0.35 proline, 0.57 serine, 0.27 threonine, 0.073 tryptophan, and 0.20 valine, 2 mM
glutamine). (IeK) Quantication of area under the curve (AUC) is shown for the various stimulatory media in females (I), males (J) and donors with T2D (K). (I) In females, insulin
secretion from ND islets was not signicantly higher than T2D islets under any culture condition (p ¼0.4806 [AAM þLG], p ¼0.2270 [AAM þHG], p ¼0.1384
[AAM þHG þIBMX], and p ¼0.1465 [KCl]; unpaired Studentst-test). (J) In males, insulin secretion from ND islets was signicantly higher than T2D islets under 4 mM
AAM þ16.7 mM glucose (HG) þ0.1 mM IBMX stimulation (p ¼0.0442 [AAM þHG þIBMX]; unpaired Studentst-test), but not in other conditions (p ¼0.5315 [AAM þLG],
p¼0.0818 [AAM þHG], and p ¼0.2259 [KCl]; unpaired Studentst-test). (K) Total insulin secretion showed a trend toward lower secretion in T2D male islets than ND male islets
(p ¼0.1514 and p ¼0.0503 for females and males, respectively; unpaired Studentst-test). *indicates p <0.05; ns indicates not signicant; error bars indicate SEM.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 5
each sex, we treated islets with thapsigargin (Tg), a specic inhibitor of
the sarcoplasmic/endoplasmic reticulum Ca
2þ
-ATPase (SERCA) that
induces ER stress and the UPR by lowering ER calcium levels [83,87].
At 2 h post-Tg treatment, protein synthesis was repressed as expected
in both male and female islet cells (Figure 3A,B; Fig. S6C). At 24 h
post-Tg treatment, we found that protein synthesis was restored to
higher-than basal levels in female islet cells, but not in male islet cells
(Figure 3A,B; Fig. S6C). Importantly, a two-way ANOVA showed that
recovery from protein synthesis repression was signicantly different
between males and females (sex:treatment interaction p <0.0001).
This suggests that while protein synthesis repression associated with
ER stress was transient in female islets, this phenotype persisted for
longer in male islets. Because insulin biosynthesis accounts for
approximately half the total protein production in
b
cells [88], one
potential explanation for the sex-specic recovery from protein syn-
thesis repression is a sex difference in transcriptional changes to in-
sulin. To test this, we quantied GFP levels in
b
cells isolated from
male and female mice with GFP knocked into the endogenous mouse
Ins2 locus (Ins2
GFP/WT
)[51,89]. While ER stress induced a signicant
reduction in Ins2 gene activity, this response was equivalent between
the sexes. This suggests Ins2 transcriptional changes cannot fully
explain the sex difference in recovery from protein synthesis repression
during ER stress (Figs. S7AeC).
Given the prolonged protein synthesis repression in males following ER
stress, we next quantied cell death, another ER stress-associated
phenotype [86], in male and female islets. Using a kinetic cell death
assay, we observed clear sex differences in Tg-induced cell death at
0.1
m
M and 1.0
m
M Tg doses throughout the time course of the
experiment (Figure 3C,D). Notably, viability prior to the assay was not
different between males and females (Fig. S8). After 84 h of Tg
treatment, no signicant increase in female islet cell apoptosis was
observed with either 0.1
m
M or 1.0
m
M Tg treatment compared with
controls (Figure 3E). In contrast, cell death was signicantly higher at
both the 0.1
m
M and the 1.0
m
M doses of Tg in male islet cells
compared with vehicle-only controls (Figure 3F). Our analysis shows
the magnitude of Tg-induced cell death was larger in male islet cells
compared with female islet cells (sex:treatment interaction p ¼0.0399
[0.1
m
M], p ¼0.0007 [1.0
m
M]). While one possible explanation for
these data is that female islets are resistant to Tg-induced cell death,
we found a signicant increase in apoptosis in both female and male
islet cells treated with 10
m
MTg(Figure 3G,H, sex:treatment inter-
action p ¼0.0996 [0.1
m
M]; data graphed separately due to different
DMSO control). This suggests female islets were more resilient to mild
ER stress caused by low-dose Tg than male islets.
To determine whether this increased ER stress resilience was caused
by differential UPR signaling, we monitored levels of several protein
markers of UPR activation including binding immunoglobulin protein
(BiP), phosphorylated inositol-requiring enzyme 1 (pIRE1), phosphor-
ylated eukaryotic initiation factor alpha (peIF2
a
), and C/EBP homolo-
gous protein (CHOP) [90,91] after treating male and female islets with
1
m
M Tg for 24 h. We found no sex difference in UPR protein markers
between male and female islets without Tg treatment (Figs. S9AeD)
and observed a signicant increase in levels of pIRE1
a
and CHOP in
islets from both sexes and BiP in female islets after a 24-hour Tg
treatment (Figs. S9AeD). Lack of a sex difference in protein markers
suggests UPR activation by Tg treatment was similar between male
and female islets at 20 weeks of age. This nding differs from the
male-biased UPR activation reported in the KINGS mouse model of
endogenous ER stress [37]. While one potential explanation for this
discrepancy is that Tg treatment induces acute ER stress in contrast to
the chronic ER stress in KINGS mice, further experiments will be
needed to conrm this possibility. Of note, we reproduced the male-
biased induction of BiP in islets isolated from 60-week-old male and
female mice (Figs. S9EeG), suggesting that age contributes to the sex
difference in UPR activation. Together, our data indicate that despite
equivalent UPR activation in male and female islets treated with Tg,
signicant sex differences exist in ER stress-associated protein syn-
thesis repression and cell death.
3.4. Female islets retain greater
b
cell function during ER stress in
mice
We next examined glucose-stimulated insulin secretion in islets
cultured under basal conditions and during Tg treatment (Figure 4A). In
all conditions tested, high glucose signicantly stimulated insulin
secretion in both sexes (Fig. S10A); however, we identied sex dif-
ferences in how well islets sustained glucose-stimulated insulin
secretion during longer Tg treatments (Figure 4B,C, Fig. S10A,B).
Female islets, in both low and high glucose, maintained robust insulin
secretion during Tg treatment (Figure 4B). Specically, we observed a
signicant increase in insulin secretion after short Tg treatment (0 and
2 h post-Tg), with a return to basal secretion levels 4 h post-Tg
(Figure 4B). Because Tg is a drug that depletes ER calcium stores, it
may induce an acute rise in cytosolic calcium that could explain this
acute increase in high glucose-stimulated insulin secretion in Tg-
treated samples compared with vehicle [83]. In contrast, male islets
showed no signicant increase in insulin secretion after short Tg
treatment, and there was a signicant drop in insulin secretion at 4 h
post-Tg treatment (Figure 4C). This suggests female islets sustained
insulin secretion for a longer period than male islets during ER stress.
Given that insulin content measurements showed insulin content
signicantly increased during the 4-hour Tg treatment in female islets,
but not male islets (Figure 4D, Fig. S10C), our data suggest one reason
female islets maintain insulin secretion during ER stress is by aug-
menting islet insulin content. Proinsulin secretion followed similar
trends to those we observed with insulin secretion (Figs. S10D,E), but
Tg treatment reduced proinsulin content to a greater degree in male
Table 1 eHuman
b
cell pathway gene numbers. The number of genes
corresponding to each T2D upregulated pathway in males, females or both
sexes.
Pathway Name Number of Pathway Genes
Unique
Male
Common Unique
Female
Asparagine N-linked glycosylation 16 9 4
Cellular responses to stimuli 49 9 6
Cellular responses to stress 47 9 6
COPI-dependent Golgi-to-ER retrograde
trafc
762
COPI-mediated anterograde transport 7 5 2
ER to Golgi Anterograde Transport 10 5 2
Golgi-to-ER retrograde transport 7 6 2
Hedgehog ligand biogenesis 12 4 1
Hh mutants abrogate ligand secretion 12 3 1
Hh mutants are degraded by ERAD 12 3 1
IRE1alpha activates chaperones 9 3 1
Metabolism of proteins 69 20 13
Signaling by Hedgehog 16 6 2
The role of GTSE1 in G2/M progression
after G2 checkpoint
14 4 1
Unfolded Protein Response (UPR) 13 3 1
XBP1(S) activates chaperone genes 8 3 1
Original Article
6MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
Figure 2: Sex-biased gene expression in mouse islet bulk RNAseq. (A) Principal component analysis (PCA) of RNAseq data from male and female mouse islets. (B) Over-
representation analysis (ORA) of all signicantly differentially expressed genes (p-adj <0.01) from male and female mouse islets. Top 30 enriched KEGG pathways (large nodes;
size ¼proportional to connections, darker red color ¼greater signicance) and associated genes (small nodes; green ¼male enriched, yellow ¼female enriched). (C) Top
signicantly enriched Reactome pathways from the top 1000 signicantly differentially expressed genes. (p-adj <0.01) for males and females. Gene ratio is calculated as k/n,
where k is the number of genes identied in each Reactome pathway, and n is the number of genes from the submitted gene list participating in any Reactome pathway. For
complete Reactome pathway lists see Supplementary le 3. (D) All transcripts of differentially expressed genes under the gene ontology term Cellular response to ER stress
(GO:0034976) and genes labeled by their role in transcription, translation, protein processing, protein folding, secretion and protein quality control. (E) All transcripts of differentially
expressed ribosomal genes.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 7
islets (Figure 4E). There was no sex difference in the ratio of
proinsulin:insulin content at any timepoint (Fig. S10G). This suggests
that in addition to a greater ability to maintain glucose-stimulated
insulin secretion during ER stress, female islets also show a larger
increase in insulin content and a smaller decrease in proinsulin
content in this context.
To determine whether female islets have improved
b
cell function under
ER stress in other contexts, we next monitored glucose-stimulated in-
sulin secretion and glucose tolerancein mice at 20 weeks, an age where
we demonstrated that insulin sensitivity was similar between the sexes
(Figure 4FeH; Fig. S4). We found that fasting plasma insulin levels were
higher in males (Figure 4F) and that the sexes showed similar glucose
Figure 3: Sex differences in mouse islet ER stress-associated phenotypes. (A, B) Protein synthesis was quantied in dispersed islet cells from 20-week-old male and female B6
mice after treatment with 1
m
M Tg for 2- or 24-hours. (A) In female islet cells, protein synthesis was signicantly lower after a 2-hour Tg treatment compared to control (p <0.0001;
one-way ANOVA followed by Tukey HSD test), signicantly higher after a 24-hour Tg treatment compared to a 2-hour Tg treatment (p <0.0001; one-way ANOVA followed by Tukey
HSD test) and recovered to a signicantly higher level than control levels p <0.0001; one-way ANOVA followed by Tukey HSD test). (B) In male islet cells, protein synthesis was
signicantly lower after a 2- and 24-hour Tg treatment compared to control (p <0.0001 for both treatments; one-way ANOVA followed by Tukey HSD test) and was not signicantly
different after a 24-hour treatment compared to a 2-hour Tg treatment p ¼0.3022; one-way ANOVA followed by Tukey HSD test). The magnitude of protein synthesis repression and
recovery was signicantly different in all sex:treatment interactions (p ¼0.0015 [DMSO-2hr], p <0.0001 [DMSO-24hr], p <0.0001 [2hre24hr]; two-way ANOVA followed by Tukey
HSD test). (CeH) Quantication of propidium iodide (PI) cell death assay of dispersed islets from 20-week-old male and female B6 mice treated with thapsigargin (0.1
m
M, 1
m
Mor
10
m
M Tg) or DMSO for 84 h n ¼4e6mice,>1000 cells per group. Percentage (%) of PI positive cells was quantied as the number of PI-positive/Hoechst 33342-positive cells in
female (C) and male (D) islet cells. Relative cell death at 84 h in Tg treatments compared with DMSO treatment in females (E, G) and males (F, H). The control for both 0.1 and
1.0
m
M Tg treatmentsis 0.1% DMSO (E, F). The control for 10
m
M Tg treatment is 0.2% DMSO (G, H). In female islet cells, cell death was signicantly higher in 10
m
MTgcomparedto
control (p <0.0001; unpaired Studentst-test). In male islet cells, cell death was signicantly higher in 0.1, 1.0 and 10
m
M Tg compared to control (p ¼0.0230 [0.1
m
M], p <0.0001
[1
m
M] and p <0.0001 [10
m
M]; unpaired Studentst-test) (D). For E-H, at 84 h the % of PI positive cells for each treatment was normalized to the DMSO control avg for each sex. *
indicates p <0.05, ** indicates p <0.01, **** indicates p <0.0001; ns indicates not signicant; error bars indicate SEM.
Original Article
8MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
Figure 4: Sex differences in ex vivo and in vivo insulin secretion. (A) Experimental workow of static glucose-stimulated insulin secretion. (B, C) Relative high glucose (20 mM;
high glucose, HG) in treatments compared with DMSO in female (B) and male (C) islets. Female islet HG secretion was signicantly higher compared with control after 0- and 2-
hour Tg pre-treatments (p ¼0.0083 [0-hour] and p ¼0.0371 [2-hour]; Mann Whitney test). Male islet HG secretion was signicantly lower compared with control after a 4-hour Tg
pre-treatment (p ¼0.0013; Mann Whitney test). (D) Insulin content. Female islet insulin content was signicantly higher compared with control after a 4-hour Tg pre-treatment
(p ¼0.0269; Mann Whitney test). (E) Proinsulin content. Female islet proinsulin content was signicantly lower compared with control after a 2-hour Tg pre-treatment (p ¼0.0437;
Mann Whitney test). Male islet proinsulin content was signicantly lower compared with control after 2- and 4-hour Tg pre-treatments (p ¼0.0014 [2-hour] and p ¼0.0005 [4-
hour]; Mann Whitney test). (FeH) Physiology measurements after a 6-hour fast in 20-week-old male and female B6 mice. (F, G) Insulin levels from glucose-stimulated insulin
secretion tests (F: nM, G: % basal insulin) following a single glucose injection (2 g glucose/kg body weight, i.p). Area under the curve (AUC) calculations (n ¼13 females, n ¼18
males). (F) Insulin levels were signicantly higher in male mice at 0 min and 30 min post injection (p ¼0.0063 [0 min] and p ¼0.0009 [30 min]; Studentst-test). AUC was
signicantly higher in males (p ¼0.0159; Studentst-test). (G) Insulin levels (% baseline). Glucose-stimulated insulin secretion was signicantly higher in female mice 15 min post
injection (p ¼0.0279; Studentst-test). (H) Glucose levels from glucose tolerance tests following a single glucose injection (2 g glucose/kg body weight). AUC calculations (n ¼11
females, n ¼11 males). For B-E, grey triangles indicate the concentration of insulin or proinsulin from ve islets, black circles indicate the average values per mouse. *indicates
p<0.05, ** indicates p <0.01, *** indicates p <0.001; ns indicates not signicant; error bars indicate SEM.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 9
tolerance (Figure 4H). To determine the magnitude of the acute glucose-
stimulated insulin secretion response in each sex, we normalized
glucose-stimulated insulin secretion to basal insulin secretion levels. We
found that females showed a trend toward higher acute glucose-
stimulated insulin secretion response in C57BL/6J mice (Figure 4G),
and signicantly higher glucose-stimulated insulin secretion in 10- and
22-week-old Insr
f/f
:Ins1Cre
/
:nTnG
þ/
mice (Figs. S11AeD;data
replotted from a prior Johnson lab study [53]). These ndings align with
data showing higher glucose-stimulated insulin secretion from prior
studies in humans and rodents [5,10,16,92,93]. We also found that
fasting blood glucose levels in female mice were more resilient to the
near-total insulin gene knockout in Ins1
/
;Ins2
/
;Pdx1CreER mice
given tamoxifen (Fig. S11E; replotted from published [54] and unpub-
lished Johnson lab data). We cannot rule out all potential factors that
may contribute to the sex differences in blood glucose levels following
near-total loss of insulin gene function (e.g. RNA stability, translation
efciency); however, given that the burden of insulin production [54]
leads to ER stress even in normal physiological conditions, our data add
further support to a model in which
b
cells in female mice show a
greater ability to maintain glucose-stimulated insulin production and
secretion during ER stress.
3.5. Sex differences in islet transcriptional and proteomic
responses to ER stress in mice
To gain insight into the differential ER stress-associated phenotypes in
male and female islets, we investigated global transcriptional changes
after either a 6- or 12-hour Tg treatment in each sex. Principal
component analysis and unsupervised clustering shows that islets
clustered by sex, treatment, and treatment time (Figure 5A; Fig. S12A).
The majority of the variance was explained by treatment (Figure 5B),
and pathway enrichment analysis conrms the UPR as the top upre-
gulated pathway in Tg-treated male and female islets at both 6- and
12-hours after treatment (Figs. S13A,B; Supplementary le 4). While
some UPR-associated genes differentially regulated by Tg treatment
were shared between the sexes (6-hour: 29/36, 12-hour: 25/31),
biological sex explained a large proportion of variance in the gene
expression response to ER stress. This suggests the transcriptional
response to ER stress was not fully shared between the sexes. Indeed,
after a 6-hour Tg treatment, 32.6% (2247/4655) of genes that were
differentially expressed between DMSO and Tg were unique to one sex
(881 to females, 1376 to males). After a 12-hour Tg treatment, 29%
(2259/7785) were unique to one sex (1017 to males, 1242 to females).
To describe the transcriptional response of each sex to Tg treatment in
more detail, we used a two-way ANOVA to identify genes that were
upregulated, downregulated, or unchanged in male and female islets
between 6- and 12-hours post-Tg (Supplementary le 5). By per-
forming pathway enrichment analysis, we were able to determine
which processes were shared, and which processes differed, between
the sexes during Tg treatment. For example, we observed a signicant
increase in mRNA levels of genes corresponding to pathways such as
cellular responses to stimuli, stress, and starvation in both male and
female islets between 6- and 12-hour Tg treatments (Figure 5C;
Supplementary le 4), suggesting Tg has similar effects on genes
related to these pathways in both sexes. Similarly, the Tg-induced
changes in mRNA levels of genes related to apoptosis were largely
shared between the sexes (Figs. S14A,B). While this suggests that the
sex-specic regulation of genes related to apoptosis does not fully
account for the susceptibility of male islets to low-dose Tg-induced cell
death, in-depth studies of
b
cell apoptosis will be needed to conrm
this point.
In contrast to these non-sex-specic changes in gene expression,
there was a male-specic increase in mRNA levels of genes associated
with translation during Tg treatment (Figure 5C; Supplementary le 4).
In females, there was a decrease in mRNA levels of genes associated
with
b
cell identity, such as Pklr,Rfx6,Hnf4a,Slc2a2,Pdx1, and MafA
(Fig. S15A), and in genes linked with regulation of gene expression in
b
cells (Figure 5C). Neither of these categories were altered between 6-
and 12-hour Tg treatments in males (Figure 5C; Fig. S15B). While our
data suggests some aspects of the gene expression response to ER
stress were shared between the sexes, we found that many genes
corresponding to important cellular processes were differentially
regulated during Tg treatment in only one sex.
Beyond sex-specic transcriptional changes following Tg treatment,
ER stress also had a sex-specic effect on the islet proteome. Although
the majority of proteins were downregulated by Tg treatment due to the
generalized repression of protein synthesis under ER stress
(Figure 5D), we identied 47 proteins (35 downregulated, 12 upre-
gulated in Tg) that were differentially expressed in female islets and 82
proteins (72 downregulated, 10 upregulated in Tg) that were differ-
entially expressed after Tg treatment in male islets (Supplementary
Table 1). Proteins downregulated only in females include proteins
associated with the GO term endoplasmic reticulum to Golgi vesicle-
mediated transport(GO:0006888) (BCAP31, COG5, COG3, GOSR1),
whereas proteins downregulated only in males include proteins
associated with GO terms insulin secretion(GO:0030073) (PTPRN2,
CLTRN, PTPRN) and lysosome pathway(KEGG) (NPC2, CTSZ, LAMP2,
PSAP, CLTA). Importantly, only seven differentially expressed proteins
were in common between the sexes (Figure 5D). This suggests that as
with our phenotypic and transcriptomic data, the proteomic response
to Tg treatment was largely not shared between the sexes.
To integrate our islet transcriptome and partial islet proteome data, we
assessed the direction of changes to mRNA and protein levels
following Tg treatment. In females, the number of differentially
expressed islet proteins with concordant mRNA changes was 43% (20/
47), whereas the number of differentially expressed islet proteins in
males with concordant mRNA changes was 49% (40/82) (Fig. S16;
Supplementary le 6). This data suggests that many genes with dif-
ferential mRNA expression during Tg treatment show congruent
changes in protein abundance. When we next asked whether the islet
genes with concordant changes in mRNA and protein levels during Tg
treatment were shared between the sexes, we found that only 7% (4/
56) of these genes were differentially expressed in both sexes during
Tg treatment (Tmem27,Emb,Fkbp9,Pdcd4). The remaining 93% (51/
55) of islet genes with concordant changes in mRNA and protein levels
during Tg treatment were differentially expressed in only one sex.
Thus, when taken together, our islet transcriptome and partial prote-
ome data suggest that male and female islets show distinct responses
to ER stress.
Of the islet genes with concordant changes in mRNA and protein levels,
several have been linked with
b
cell and/or islet function. For example,
Tmem27 plays a role in enhancing GSIS [94], and Pdcd4 expression is
associated with
b
cell death under stressed conditions [95,96]. Atp6ap2
and Lamp2 have important roles in autophagy [97e99], Ptprn2 is
required for the accumulation of insulin granules [100], and both Ptprn2
and Chga are involved in glucose-stimulated insulin secretion
[101,102]. Chga is also important for maintaining islet volume and islet
cell composition [103], and studies show that Tbc1d1 inuences insulin
secretion and
b
cell mass in rodents [104,105]. Because the Tbc1d1
study [104,105], and others [94e103], used single- or mixed-sex
models, or cell lines, future studies will need to address whether
these effects on
b
cell and/or islet function are shared between the
Original Article
10 MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
Figure 5: Sex-specic transcriptomic and proteomic proles following ER stress in mouse islets. (A) Principal component analysis (PCA) of RNAseq data from male and
female mouse islets treated with DMSO or 1
m
M Tg for 6- or 12-hours. (B) Spearman correlation depicting the variance for the rst 5 principal components. (C) Top signicantly
enriched Reactome pathways from the top 1000 signicantly differentially expressed genes (p-adj<0.01) for females and males that were upregulated or downregulated between
6 and 12 h of Tg treatment. Gene ratio is calculated as k/n, where k is the number of genes identied in each Reactome pathway, and n is the number of genes from the submitted
gene list participating in any Reactome pathway. (D) Protein abundance from proteomics data of female and male mouse islets treated with DMSO or 1
m
M Tg for 6 h. Top 45
differentially expressed proteins are shown (p <0.05).
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 11
sexes. Additional studies will also be needed on genes identied in our
analysis that were not previously linked with
b
cell and/or islet function
(Supplementary le 6). This will elucidate whether the sex-specic
regulation of mRNA and protein levels during Tg treatment affects
b
cell and/or islet function in either sex.
4. DISCUSSION
Emerging evidence shows biological sex affects many aspects of
b
cell
gene expression and function. Yet, many studies on
b
cells do not
include both sexes, or fail to analyze male and female data separately.
To address this gap in knowledge, the goal of our study was to provide
detailed information on sex differences in islet and
b
cell gene
expression and function in multiple contexts. In humans, we used a
large scRNAseq dataset from ND and T2D donors to reveal signicant
male-female differences in the magnitude of gene expression changes,
and in the identity of genes that were differentially regulated, between
ND and T2D donors. While these data do not address the reasons for the
sex-biased risk of T2D, our ndings suggest
b
cell gene expression
changes in T2D are not fully shared between the sexes. These data
provide a useful resource to support future studies on how medication,
disease progression, age, or body mass index contribute to the sex
difference in
b
cell gene expression in T2D. Sex-based analysis of
human
b
cell gene expression data will also clarify the mechanisms
underlying our nding that
b
cells from female donors with T2D
maintain higher insulin production than male donors with T2D.
In mice, we generated a large RNAseq dataset using islets isolated from
20-week-old males and females. We used an ITT to show that male
and female mice have similar insulin sensitivity at this age. We
acknowledge that the ITT may not be sensitive enough to pick up small
differences, so future studies will need to use a hyperinsulinemic-
euglycemic clamp to further compare insulin sensitivity between the
sexes. Despite this potential limitation, our unbiased analysis of gene
expression in islets from males and females revealed sex differences in
genes associated with the UPR under normal physiological conditions.
This differential gene expression was signicant, as female islets were
more resilient to phenotypes caused by ER stress and UPR activation
than male islets, showed sex-specic transcriptional and proteomic
changes in this context, and had a greater ability to maintain glucose-
stimulated insulin production and secretion during ER stress. Collec-
tively, these data suggest that in rodents,
b
cells from females are more
resilient to ER stress. Considering the well-established links between
ER stress and T2D [90,106e108], our data suggests a model in which
female
b
cells have a greater ability to maintain glucose-stimulated
insulin secretion in T2D because they are more resilient to ER stress
and UPR activation. While future studies are needed to test this working
model, and to assess the relative contribution of sex differences in
b
cells to the sex-biased risk of T2D, our ndings highlight the impor-
tance of including both sexes in islet and
b
cell studies.
Including both sexes in our analysis of
b
cell gene expression in human
ND and T2D allowed us to uncover genes that were differentially
regulated in T2D in each sex. Because many of these genes may have
been missed if the scRNAseq data was not analyzed by sex, our ndings
advance knowledge of
b
cell changes in T2D by identifying additional
genes that are differentially regulated in this context. This knowledge
adds to a growing number of studies that identify sex differences in
b
cell gene expression during aging in humans [12], and in mice fed either
anormal[4,11] or a high fat diet [11]. Further, given that our RNAseq on
islets from male and female mice with similar insulin sensitivity identies
genes and biological pathways that align with previous studies on sex
differences in murine
b
cell gene expression [4,11], our data suggests
that sex differences in islet and
b
cell gene expression cannot be
explained solely by a male-female difference in peripheral insulin
resistance. Instead, there is likely a basal sex difference in
b
cell gene
expression that forms the foundation for sex-specic transcriptional
responses to perturbations such as ER stress and T2D. By generating
large islet gene expression datasets from male and female mice with
similar peripheral insulin sensitivity and from islets subjected to phar-
macological induction of ER stress, our studies provide a foundation of
knowledge for future studies aimed at studying the causes and con-
sequences of sex differences in islet ER stress responses and
b
cell
function following UPR activation. This will provide deeper mechanistic
insight into the sex-specic phenotypic effects reported in animal
models of
b
cell dysfunction [35e39,109e112] and the sex-biased risk
of diseases such as T2D that are associated with
b
cell dysfunction
[12,22,113,114].
Beyond gene expression,our sex-based analysis of mouse islets allowed
us to uncover male-female differences in ER stress-associated pheno-
types (e.g. protein synthesis repression, cell death). While previous
studies identify a sex difference in
b
cell loss in diabetic mouse models
[37,39,115], and show that estrogen plays a protective role via estrogen
receptor
a
(ER
a
) against ER stress to preserve
b
cell mass and prevent
apoptosis in cell lines, mouse models, and human islets [39,115,116],
we extend prior ndings by showing that differences in ER stress-
induced cell death were present in the context of similar insulin sensi-
tivity between the sexes. This suggests sex differences in ER stress-
associated phenotypes do not solely depend on male-female differ-
ences in peripheral insulin sensitivity. Indeed, islets isolated from males
and females with similar insulin sensitivity also showa sex difference in
protein synthesis repression, a classical ER stress-associated phenotype
[86]. While estrogen affects insulin biosynthesis via ER
a
[117], future
studies will need to determine whetherestrogen also allows female islets
to restore protein synthesis to basal levels faster than male islets
following ER stress. We currentlylack this knowledge, as most studies on
UPR-mediated recovery from protein translation repression use single-
and mixed-sex animal groups, or cultured cells [118e123].
Assessing whether the recovery of protein synthesis contributes to
reduced cell death in female islets following ER stress will also be an
important task for future studies, as prior work suggests the inability to
recover from protein synthesis repression increases ER-stress induced
apoptosis [118]. Ideally, this type of study would also monitor the
activity of pathways known to regulate protein synthesis repression
during ER stress. For example, while we did not detect any changes in
levels of phosphorylated eIF2
a
(also known as Eif2s1), which is known
to mediate UPR-induced protein synthesis repression [86], our chosen
timepoints did not overlap with the rapid changes in phospho-eIF2
a
following ER stress published in other studies [124,125]. A more
detailed time course will therefore be necessary to assess p-eIF2
a
levels during ER stress in both sexes, and to test a role for phospho-
eIF2
a
in mediating differences in protein synthesis repression. More
work will also be needed to determine whether males and females
differ in
b
cell replication [126], another ER stress-related phenotype.
Ultimately, a better understanding of sex differences in ER stress-
associated phenotypes in
b
cells will provide a mechanistic explana-
tion for the strongly male-biased onset of diabetes-like phenotypes in
mouse models of
b
cell ER stress (e.g. Akita, KINGS, Munich mice)
[37,38,109]. Given the known relationship between ER stress,
b
cell
death, and T2D, studies on the male-female difference in
b
cell ER
stress-associated phenotypes may also advance our understanding of
the male-biased risk of developing T2D in some population groups.
Original Article
12 MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
A further benet of additional studies on the sex difference in
b
cell ER
stress responses will be to identify mechanisms that support
b
cell
insulin production. In rodents, we found that female islets maintained
high glucose-stimulated insulin secretion and increased insulin content
following ER stress, whereas male islets showed signicant repression
of high glucose-stimulated insulin secretion under the same condi-
tions. In humans, while a study using a mixed-sex group of T2D donors
shows
b
cells experience ER stress associated with
b
cell dysfunction
[63], we found that changes to
b
cell insulin secretion in T2D were not
the same between the sexes. Specically, the magnitude of the
reduction in insulin release by
b
cells from female donors with T2D
was smaller than in
b
cells from male donors with T2D. Together with
our data from rodents, this suggests female
b
cells maintain enhanced
insulin production and/or secretion in multiple contexts, and the
increased
b
cell function cannot be solely attributed to a sex difference
in peripheral insulin sensitivity.
Clues into potential ways that female
b
cells maintain improved insulin
production and secretion emerge from our examination of the tran-
scriptional response to ER stress in mice of each sex. Our data shows
that Tg treatment induces gene expression changes characteristic of
ER stress [127], and revealed similar biological pathways that were
upregulated in T2D donors. Furthermore, we identied signicant
differences between male and female islets in the transcriptional
response to ER stress over time. One notable nding was that a greater
number of
b
cell identity genes were downregulated between 6- and
12-hour Tg treatments in females, but not in males. Because most
studies on the relationship between
b
cell identity and function used a
mixed-sex pool of islets and
b
cells [79,128,129], more studies will be
needed to test whether there are sex-specic changes to
b
cell identity
during ER stress, and to determine the functional consequences of this
sex-specic effect.
Overall, our data demonstrates sex differences in
b
cell function in
multiple contexts. One potential explanation for these differences is the
sex-specic regulation of
b
cell ER stress responses and function.
Indeed, sex differences in ER stress and protein markers of apoptosis
were observed in mouse kidney cells [130], suggesting that studying
b
cell ER stress may provide insight into this difference in other cell
types. Alternatively, it is possible that there are sex differences in
b
cell
number that account for the male-female differences in
b
cell function
that we observe. For example, considering two published studies
indicate males have fewer
b
cells [6,37], the burden of maintaining
glucose homeostasis may fall on a smaller number of cells in males,
leading to higher susceptibility to ER stress. Future studies will need to
address sex differences in
b
cell number relative to pancreas size and
body size to test this possibility. Ultimately, a better understanding of
changes to
b
cell gene expression and function in males and females
will suggest effective ways to reverse disease-associated changes to
this important cell type in each sex, improving equity in health out-
comes [131].
4.1. Conclusions
Our study reports signicant sex differences in islet and
b
cell gene
expression and stress responses in both humans and mice. These
differences likely contribute to sex differences in
b
cell resilience,
allowing female
b
cells to show a greater ability to maintain glucose-
stimulated insulin production and secretion across multiple contexts.
This knowledge forms a foundation for future studies aimed at un-
derstanding how sex differences in
b
cell function affect physiology
and the pathophysiology of diseases such as T2D.
AUTHOR CONTRIBUTIONS
G. P. B. conceived studies, conducted experiments, interpreted ex-
periments, wrote the manuscript
Y. X. performed bioinformatic analysis and data visualization
J. C. created custom R scripts (single-cell GFP tracking)
S. W. analyzed data (human RNAseq)
C. C. created custom R scripts (mouse RNAseq analysis)
J. A. Z. analyzed data (HPAP perifusions)
S. S. conducted experiments (in vivo physiology)
E. P. conducted experiments (islet western blots)
X. H. conducted experiments (dissections)
J. D. J. conceived studies, interpreted experiments, edited the
manuscript
E. J. R. conceived studies, interpreted experiments, edited the
manuscript, and is the guarantor of this work
FUNDING
This study was supported by operating grants to E.J.R. from the
Michael Smith Foundation for Health Research (16876), Canadian In-
stitutes of Health Research (GS4-171365), the Canadian Foundation
for Innovation (JELF-34879), and Diabetes Canada (OG-3-22-5646-
ER), and to J.D.J. (PJT-152999) from the Canadian Institutes of Health
Research, and core support from the JDRF Centre of Excellence at UBC
(3-COE-2022-1103-M-B). J.D.J. was funded by a Diabetes Investi-
gator Award from Diabetes Canada.
DATA AVAILABILITY
Details of all statistical tests and p-values as well as the raw data are
provided in Supplementary les. Supplementary les are available as
Brownrigg, George (2023), Sex differences in islet stress responses
support female
b
cell resilience, Mendeley Data, V1, doi: https://doi.
org/10.17632/ftcs6xj9ft.1. RNAseq data is available at PRJNA842443
and PRJNA842371.
ACKNOWLEDGEMENTS
We thank members of the Rideout and Johnson lab for valuable feedback. We thank
our animal care staff for supporting our animal husbandry, UBC Biomedical Research
Center for performing RNA sequencing, and UVic Genome BC Proteomics Center for
performing proteomics. We acknowledge that our research takes place on the
traditional, ancestral, and unceded territory of the Musqueam people; a privilege for
which we are grateful.
CONFLICT OF INTEREST
The authors declare no competing interest.
APPENDIX A. SUPPLEMENTARY DATA
Supplementary data to this article can be found online at https://doi.org/10.1016/j.
molmet.2023.101678.
REFERENCES
[1] Parchami A, Kusha S. Effect of sex on histomorphometric properties of
Langerhans islets in native chickens. Vet Res Forum 2015;6(4):327e30.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 13
[2] Rideout EJ, Narsaiya MS, Grewal SS. The sex determination gene transformer
regulates male-female differences in Drosophila body size. PLoS Genet 2015
Dec 28;11(12):e1005683.
[3] Millington JW, Brownrigg GP, Chao C, Sun Z, Basner-Collins PJ, Wat LW,
et al. Female-biased upregulation of insulin pathway activity mediates the sex
difference in Drosophila body size plasticity. Elife 2021 Jan 15;10:e58341.
[4] Stancill JS, Osipovich AB, Cartailler JP, Magnuson MA. Transgene-associated
human growth hormone expression in pancreatic
b
-cells impairs identica-
tion of sex-based gene expression differences. Am J Physiol Endocrinol
Metab 2019 Feb 1;316(2):E196e209.
[5] Horie I, Abiru N, Eto M, Sako A, Akeshima J, Nakao T, et al. Sex differences in
insulin and glucagon responses for glucose homeostasis in young healthy
Japanese adults. J Diabetes Investig 2018 Nov;9(6):1283e7.
[6] Marchese E, Rodeghier C, Monson RS, McCracken B, Shi T, Schrock W, et al.
Enumerating
b
-cells in whole human islets: sex differences and associations
with clinical outcomes after islet transplantation. Diabetes Care 2015 Nov
1;38(11):e176e7.
[7] Oliva M, Muñoz-Aguirre M, Kim-Hellmuth S, Wucher V, Gewirtz ADH,
Cotter DJ, et al. The impact of sex on gene expression across human tissues.
Science 2020 Sep 11;369(6509):eaba3066.
[8] Enge M, Arda HE, Mignardi M, Beausang J, Bottino R, Kim SK, et al. Single-
cell analysis of human pancreas reveals transcriptional signatures of aging
and somatic mutation patterns. Cell 2017 Oct 5;171(2):321e330.e14.
[9] Schaum N, Lehallier B, Hahn O, Pálovics R, Hosseinzadeh S, Lee SE, et al.
Aging hallmarks exhibit organ-specic temporal signatures. Nature 2020
Jul;583(7817):596e602.
[10] Hall E, Volkov P, Dayeh T, Esguerra JLS, Salö S, Eliasson L, et al. Sex dif-
ferences in the genome-wide DNA methylation pattern and impact on gene
expression, microRNA levels and insulin secretion in human pancreatic islets.
Genome Biol 2014 Dec 3;15(12):522.
[11] Liu G, Li Y, Zhang T, Li M, Li S, He Q, et al. Single-cell RNA sequencing
reveals sexually dimorphic transcriptome and type 2 diabetes genes in
mouse islet
b
cells. Dev Reprod Biol 2021 Jun 1;19(3):408e22.
[12] Arrojoe Drigo R, Erikson G, Tyagi S, Capitanio J, Lyon J, Spigelman AF, et al.
Aging of human endocrine pancreatic cell types is heterogeneous and sex-
specic. bioRxiv 2019 Aug;729541.
[13] Li T, Jiao W, Li W, Li H. Sex effect on insulin secretion and mitochondrial
function in pancreatic beta cells of elderly Wistar rats. Endocr Res 2016 Jul
2;41(3):167e79.
[14] Tura A, Pacini G, Moro E, Vrbíková J, Bendlová B, Kautzky-Willer A. Sex- and
age-related differences of metabolic parameters in impaired glucose meta-
bolism and type 2 diabetes compared to normal glucose tolerance. Diabetes
Res Clin Pract 2018 Dec 1;146:67e75.
[15] Kautzky-Willer A, Brazzale AR, Moro E, Vrbíková J, Bendlova B,
Sbrignadello S, et al. Inuence of increasing BMI on insulin sensitivity and
secretion in normotolerant men and women of a wide age span. Obesity
2012;20(10):1966e73.
[16] Basu R, Man CD, Campioni M, Basu A, Klee G, Toffolo G, et al. Effects of age
and sex on postprandial glucose metabolism: differences in glucose turnover,
insulin secretion, insulin action, and hepatic insulin extraction. Diabetes 2006
Jul 1;55(7):2001e14.
[17] Nuutila P, Knuuti MJ, Mäki M, Laine H, Ruotsalainen U, Teräs M, et al. Gender
and insulin sensitivity in the heart and in skeletal muscles: studies using
positron emission tomography. Diabetes 1995 Jan 1;44(1):31e6.
[18] Lundsgaard AM, Kiens B. Gender differences in skeletal muscle substrate
metabolism emolecular mechanisms and insulin sensitivity. Front Endo-
crinol 2014 Nov 13;5:195.
[19] Borissova AM, Tankova T, Kirilov G, Koev D. Gender-dependent effect of
ageing on peripheral insulin action. Int J Clin Pract 2005;59(4):422e6.
[20] Geer EB, Shen W. Gender differences in insulin resistance, body composition,
and energy balance. Gend Med 2009 Jan 1;6:60e75.
[21] Færch K, Borch-Johnsen K, Vaag A, Jørgensen T, Witte DR. Sex differences in
glucose levels: a consequence of physiology or methodological convenience?
The Inter99 study. Diabetologia 2010 May 1;53(5):858e65.
[22] Gannon M, Kulkarni RN, Tse HM, Mauvais-Jarvis F. Sex differences under-
lying pancreatic islet biology and its dysfunction. Mol Metabol 2018 Sep;15:
82e91.
[23] Tramunt B, Smati S, Grandgeorge N, Lenfant F, Arnal JF, Montagner A, et al.
Sex differences in metabolic regulation and diabetes susceptibility [Internet]
Diabetologia 2019 Nov 21. https://doi.org/10.1007/s00125-019-05040-3
[cited 2019 Dec 4].
[24] Macotela Y, Boucher J, Tran TT, Kahn CR. Sex and depot differences in
adipocyte insulin sensitivity and glucose metabolism. Diabetes 2009
Apr;58(4):803e12.
[25] Rudnicki M, Abdifarkosh G, Rezvan O, Nwadozi E, Roudier E, Haas TL. Fe-
male Mice Have Higher Angiogenesis in Perigonadal Adipose Tissue Than
Males in Response to High-Fat Diet. Front Physiol 2018;9:1452.
[26] Millington JW, Biswas P, Chao C, Xia YH, Wat LW, Brownrigg GP, et al. A low
sugar diet enhances Drosophila body size in males and females via sex-
specic mechanisms. Development 2022 Feb 23:200491. dev.
[27] Mank JE, Rideout EJ. Developmental mechanisms of sex differences: from
cells to organisms. Development 2021 Oct 14;148(19):dev199750.
[28] Oster RT, Johnson JA, Hemmelgarn BR, King M, Balko SU, Svenson LW, et al.
Recent epidemiologic trends of diabetes mellitus among status Aboriginal
adults. CMAJ (Can Med Assoc J) 2011 Sep 6;183(12):E803e8.
[29] Dyck R, Osgood N, Lin TH, Gao A, Stang MR. Epidemiology of diabetes
mellitus among First Nations and non-First Nations adults. CMAJ (Can Med
Assoc J) 2010 Feb 23;182(3):249e56.
[30] Zhou B, Lu Y, Hajifathalian K, Bentham J, Cesare MD, Danaei G, et al.
Worldwide trends in diabetes since 1980: a pooled analysis of 751
population-based studies with 4$4 million participants. Lancet 2016 Apr
9;387(10027):1513e30.
[31] Kautzky-Willer A, Harreiter J, Pacini G. Sex and gender differences in risk,
pathophysiology and complications of type 2 diabetes mellitus. Endocr Rev
2016 Jun;37(3):278e316.
[32] Heise L, Greene ME, Opper N, Stavropoulou M, Harper C, Nascimento M,
et al. Gender inequality and restrictive gender norms: framing the challenges
to health. Lancet 2019 Jun 15;393(10189):2440e54.
[33] Kautzky-Willer A, Harreiter J. Sex and gender differences in therapy of type 2
diabetes. Diabetes Res Clin Pract 2017 Sep;131:230e41.
[34] Corsetti JP, Sparks JD, Peterson RG, Smith RL, Sparks CE. Effect of dietary
fat on the development of non-insulin dependent diabetes mellitus in obese
Zucker diabetic fatty male and female rats. Atherosclerosis 2000 Feb;148(2):
231e41.
[35] Paik SG, Michelis MA, Kim YT, Shin S. Induction of insulin-dependent dia-
betes by streptozotocin. Inhibition by estrogens and potentiation by andro-
gens. Diabetes 1982 Aug;31(8 Pt 1):724e9.
[36] Verchere CB, DAlessio DA, Palmiter RD, Weir GC, Bonner-Weir S, Baskin DG,
et al. Islet amyloid formation associated with hyperglycemia in transgenic
mice with pancreatic beta cell expression of human islet amyloid polypeptide.
Proc Natl Acad Sci U S A 1996 Apr 16;93(8):3492e6.
[37] Austin ALF, Daniels Gatward LF, Cnop M, Santos G, Andersson D,
Sharp S, et al. The KINGS ins2þ/G32S mouse: a novel model of
b
-cell
endoplasmic reticulum stress and human diabetes. Diabetes 2020
Dec;69(12):2667e77.
[38] Yoshioka M, Kayo T, Ikeda T, Koizuni A. A novel locus, Mody4, distal to
D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese
C57BL/6 (akita) mutant mice. Diabetes 1997 May 1;46(5):887e94.
[39] May CL, Chu K, Hu M, Ortega CS, Simpson ER, Korach KS, et al. Estrogens
protect pancreatic
b
-cells from apoptosis and prevent insulin-decient
diabetes mellitus in mice. Proc Natl Acad Sci U S A 2006 Jun
13;103(24):9232.
Original Article
14 MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
[40] Cohrs CM, Panzer JK, Drotar DM, Enos SJ, Kipke N, Chen C, et al.
Dysfunction of persisting
b
cells is a key feature of early type 2 diabetes
pathogenesis. Cell Rep 2020 Apr 7;31(1):107469.
[41] Saisho Y.
b
-cell dysfunction: its critical role in prevention and management of
type 2 diabetes. World J Diabetes 2015 Feb 15;6(1):109e24.
[42] Segerstolpe Å, Palasantza A, Eliasson P, Andersson EM, Andréasson AC,
Sun X, et al. Single-cell transcriptome proling of human pancreatic islets in
health and type 2 diabetes. Cell Metabol 2016 Oct 11;24(4):593e607.
[43] Xin Y, Kim J, Okamoto H, Ni M, Wei Y, Adler C, et al. RNA sequencing of
single human islet cells reveals type 2 diabetes genes. Cell Metabol 2016 Oct
11;24(4):608e15.
[44] Avrahami D, Wang YJ, Schug J, Feleke E, Gao L, Liu C, et al. Single-cell
transcriptomics of human islet ontogeny denes the molecular basis of
b
-cell
dedifferentiation in T2D. Mol Metabol 2020 Jul 30;42:101057.
[45] Lawlor N, George J, Bolisetty M, Kursawe R, Sun L, Sivakamasundari V, et al.
Single-cell transcriptomes identify human islet cell signatures and reveal cell-
typeespecic expression changes in type 2 diabetes. Genome Res 2017 Feb
1;27(2):208e22.
[46] Baron M, Veres A, Wolock SL, Faust AL, Gaujoux R, Vetere A, et al. A single-
cell transcriptomic map of the human and mouse pancreas reveals inter- and
intra-cell population structure. Cell Systems 2016 Oct 26;3(4):346e360.e4.
[47] Muraro MJ, Dharmadhikari G, Grün D, Groen N, Dielen T, Jansen E, et al.
A single-cell transcriptome atlas of the human pancreas. Cell Systems 2016
Oct 26;3(4):385e394.e3.
[48] Deng S, Vatamaniuk M, Huang X, Doliba N, Lian MM, Frank A, et al.
Structural and functional abnormalities in the islets isolated from type 2
diabetic subjects. Diabetes 2004 Mar 1;53(3):624e32.
[49] Wu M, Lee MYY, Bahl V, Traum D, Schug J, Kusmartseva I, et al. Single-cell
analysis of the human pancreas in type 2 diabetes using multi-spectral
imaging mass cytometry. Cell Rep 2021 Nov 2;37(5):109919.
[50] Salvalaggio PRO, Deng S, Ariyan CE, Millet I, Zawalich WS, Basadonna GP,
et al. Islet ltration: a simple and rapid new purication procedure that avoids
coll and improves islet mass and function. Transplantation 2002 Sep
27;74(6):877e9.
[51] Chu CMJ, Modi H, Ellis C, Krentz NAJ, Skovsø S, Zhao YB, et al. Dynamic Ins2
gene activity denes
b
-cell maturity states. Diabetes 2022 Dec 1;71(12):
2612e31.
[52] Truchan NA, Brar HK, Gallagher SJ, Neuman JC, Kimple ME. A single-islet
microplate assay to measure mouse and human islet insulin secretion. Islets
2015 Oct 9;7(3):e1076607.
[53] Skovsø S, Panzhinskiy E, Kolic J, Cen HH, Dionne DA, Dai XQ, et al. Beta-cell
specic Insr deletion promotes insulin hypersecretion and improves glucose
tolerance prior to global insulin resistance. Nat Commun 2022 Feb 8;13(1):735.
[54] Szabat M, Page MM, Panzhinskiy E, Skovsø S, Mojibian M, Fernandez-
Tajes J, et al. Reduced insulin production relieves endoplasmic reticulum
stress and induces
b
cell proliferation. Cell Metabol 2016 Jan 12;23(1):
179e93.
[55] Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR:
ultrafast universal RNA-seq aligner. Bioinformatics 2013 Jan 1;29(1):15e21.
[56] Love MI, Huber W, Anders S. Moderated estimation of fold change and
dispersion for RNA-seq data with DESeq2. Genome Biol 2014 Dec
5;15(12):550.
[57] Gillespie M, Jassal B, Stephan R, Milacic M, Rothfels K, Senff-Ribeiro A, et al.
The reactome pathway knowledgebase 2022. Nucleic Acids Res 2022 Jan
7;50(D1):D687e92.
[58] Zhou G, Soufan O, Ewald J, Hancock REW, Basu N, Xia J. NetworkAnalyst
3.0: a visual analytics platform for comprehensive gene expression proling
and meta-analysis. Nucleic Acids Res 2019 Jul 2;47(W1):W234e41.
[59] Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The
Perseus computational platform for comprehensive analysis of (prote)omics
data. Nat Methods 2016 Sep;13(9):731e40.
[60] Wei T, Viliam S. R package corrplot: visualization of a correlation matrix
(version 0.92) [Internet]. 2021 [cited 2022 Dec 11]. Available from: https://
taiyun.github.io/corrplot/.
[61] Wang S, Flibotte S, Camunas-Soler J, MacDonald PE, Johnson JD. A new
hypothesis for type 1 diabetes risk: the at-risk allele at rs3842753 associates
with increased beta-cell INS messenger RNA in a meta-analysis of single-cell
RNA-sequencing data. Can J Diabetes 2021 Dec 1;45(8):775e784.e2.
[62] Hashemitabar M, Bahramzadeh S, Saremy S, Nejaddehbashi F. Glucose plus
metformin compared with glucose alone on
b
-cell function in mouse
pancreatic islets. Biomed Rep 2015 Sep;3(5):721e5.
[63] Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, et al. The
endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients.
Diabetologia 2007 Dec 1;50(12):2486e94.
[64] Lupi R, Del Guerra S, Fierabracci V, Marselli L, Novelli M, Patanè G, et al.
Lipotoxicity in human pancreatic islets and the protective effect of metformin.
Diabetes 2002 Feb;51(Suppl 1):S134e7.
[65] Kanda Y, Shimoda M, Hamamoto S, Tawaramoto K, Kawasaki F,
Hashiramoto M, et al. Molecular mechanism by which pioglitazone preserves
pancreatic
b
-cells in obese diabetic mice: evidence for acute and chronic
actions as a PPAR
g
agonist. Am J Physiol Endocrinol Metab 2010
Feb;298(2):E278e86.
[66] Murao N, Yokoi N, Takahashi H, Hayami T, Minami Y, Seino S. Increased
glycolysis affects
b
-cell function and identity in aging and diabetes. Mol
Metabol 2022 Jan;55:101414.
[67] Mizukami H, Takahashi K, Inaba W, Osonoi S, Kamata K, Tsuboi K, et al. Age-
associated changes of islet endocrine cells and the effects of body mass
index in Japanese. J Diabetes Investig 2014 Feb 12;5(1):38e47.
[68] Yinhui HE, Haiyan XU, Qi FU, Tao Y. Effects of glycosylated hemoglobin and
disease course on islet
b
-cell function in patients with type 2 diabetes. Nan
Fang Yi Ke Da Xue Xue Bao 2019 Sep 30;39(9):1003e8.
[69] Kaestner KH, Powers AC, Naji A, Consortium HPAP, Atkinson MA. NIH
initiative to improve understanding of the pancreas, islet, and autoimmunity
in type 1 diabetes: the human pancreas analysis Program (HPAP). Diabetes
2019 Jul;68(7):1394e402.
[70] Hellman B, Idahl LÅ, Lernmark Å, Täljedal IB. The pancreatic
b
-cell recog-
nition of insulin secretagogues: does cyclic AMP mediate the effect of
glucose? Proc Natl Acad Sci USA 1974 Sep;71(9):3405e9.
[71] Yang X, Schadt EE, Wang S, Wang H, Arnold AP, Ingram-Drake L, et al.
Tissue-specic expression and regulation of sexually dimorphic genes in
mice. Genome Res 2006 Aug 1;16(8):995e1004.
[72] Lu T, Mar JC. Investigating transcriptome-wide sex dimorphism by multi-level
analysis of single-cell RNA sequencing data in ten mouse cell types. Biol Sex
Differ 2020 Nov 5;11(1):61.
[73] Cottet-Dumoulin D, Lavallard V, Lebreton F, Wassmer CH, Bellofatto K,
Parnaud G, et al. Biosynthetic activity differs between islet cell types and in
beta cells is modulated by glucose and not by secretion. Endocrinology 2020
Dec 25;162(3):bqaa239.
[74] Li N, Yang Z, Li Q, Yu Z, Chen X, Li JC, et al. Ablation of somatostatin cells
leads to impaired pancreatic islet function and neonatal death in rodents. Cell
Death Dis 2018 Jun 7;9(6):1e12.
[75] Webb GC, Dey A, Wang J, Stein J, Milewski M, Steiner DF. Altered proglu-
cagon processing in an
a
-cell line derived from prohormone convertase 2
null mouse islets. J Biol Chem 2004 Jul 23;279(30):31068e75.
[76] Garcia-Barrado MJ, Ravier MA, Rolland JF, Gilon P, Nenquin M, Henquin JC.
Inhibition of protein synthesis sequentially impairs distinct steps of stimulus-
secretion coupling in pancreatic
b
cells. Endocrinology 2001 Jan 1;142(1):
299e307.
[77] Scheuner D, Mierde DV, Song B, Flamez D, Creemers JWM, Tsukamoto K,
et al. Control of mRNA translation preserves endoplasmic reticulum function
in beta cells and maintains glucose homeostasis. Nat Med 2005 Jul;11(7):
757e64.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 15
[78] Izumi T, Yokota-Hashimoto H, Zhao S, Wang J, Halban PA, Takeuchi T.
Dominant negative pathogenesis by mutant proinsulin in the Akita diabetic
mouse. Diabetes 2003 Feb;52(2):409e16.
[79] Xin Y, Dominguez Gutierrez G, Okamoto H, Kim J, Lee AH, Adler C, et al.
Pseudotime ordering of single human
b
-cells reveals states of insulin pro-
duction and unfolded protein response. Diabetes 2018;67(9):1783e94.
[80] Lipson KL, Fonseca SG, Ishigaki S, Nguyen LX, Foss E, Bortell R, et al.
Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic
reticulum-resident protein kinase IRE1. Cell Metabol 2006 Sep;4(3):245e54.
[81] Teodoro T, Odisho T, Sidorova E, Volchuk A. Pancreatic
b
-cells depend on
basal expression of active ATF6
a
-p50 for cell survival even under nonstress
conditions. Am J Physiol Cell Physiol 2012 Apr;302(7):C992e1003.
[82] Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, et al.
Translational control is required for the unfolded protein response and in vivo
glucose homeostasis. Mol Cell 2001 Jun;7(6):1165e76.
[83] Luciani DS, Gwiazda KS, Yang TLB, Kalynyak TB, Bychkivska Y, Frey MHZ,
et al. Roles of IP3R and RyR Ca2þchannels in endoplasmic reticulum stress
and
b
-cell death. Diabetes 2009 Feb 1;58(2):422e32.
[84] Riggs AC, Bernal-Mizrachi E, Ohsugi M, Wasson J, Fatrai S, Welling C, et al.
Mice conditionally lacking the Wolfram gene in pancreatic islet beta cells
exhibit diabetes as a result of enhanced endoplasmic reticulum stress and
apoptosis. Diabetologia 2005 Nov 1;48(11):2313e21.
[85] Li J, Lee B, Lee AS. Endoplasmic reticulum stress-induced apoptosis: mul-
tiple pathways and activation of p53-up-regulated modulator of apoptosis
(PUMA) and NOXA by p53. J Biol Chem 2006 Mar 17;281(11):7260e70.
[86] Sharma RB, Landa-Galván HV, Alonso LC. Living dangerously: protective and
harmful ER stress responses in pancreatic
b
-cells. Diabetes 2021
Nov;70(11):2431.
[87] Gwiazda KS, Yang TLB, Lin Y, Johnson JD. Effects of palmitate on ER and
cytosolic Ca2þhomeostasis in
b
-cells. Am J Physiol Endocrinol Metab 2009
Apr;296(4):E690e701.
[88] Fonseca SG, Gromada J, Urano F. Endoplasmic reticulum stress and
pancreatic beta cell death. Trends Endocrinol Metabol 2011 Jul;22(7):
266e74.
[89] Wakae-Takada N, Xuan S, Watanabe K, Meda P, Leibel RL. Molecular basis
for the regulation of islet beta cell mass in mice: the role of E-cadherin.
Diabetologia 2013 Apr;56(4):856e66.
[90] Shrestha N, Franco ED, Arvan P, Cnop M. Pathological
b
-cell endoplasmic
reticulum stress in type 2 diabetes: current evidence. Front Endocrinol
2021;12:650158.
[91] Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum
stress. Cell Death Differ 2004 Apr;11(4):381e9.
[92] Li T, Jiao W, Li W, Li H. Sex effect on insulin secretion and mitochondrial
function in pancreatic beta cells of elderly Wistar rats. Endocr Res 2016
Aug;41(3):167e79.
[93] Basu A, Dube S, Basu R. Men are from mars, women are from venus: sex
differences in insulin action and secretion [cited 2019 Nov 11]. In: Mauvais-
Jarvis F, editor. Sex and gender factors affecting metabolic homeostasis,
diabetes and obesity [Internet]. Cham: Springer International Publishing;
2017. p. 53e64. Available from: https://doi.org/10.1007/978-3-319-70178-
3_4 (Advances in Experimental Medicine and Biology).
[94] Altirriba J, Gasa R, Casas S, Ramírez-Bajo MJ, Ros S, Gutierrez-Dalmau A,
et al. The role of transmembrane protein 27 (TMEM27) in islet physiology and
its potential use as a beta cell mass biomarker. Diabetologia 2010 Jul
1;53(7):1406e14.
[95] Kumar S, Marriott CE, Alhasawi NF, Bone AJ, Macfarlane WM. The role of
tumour suppressor PDCD4 in beta cell death in hypoxia. PLoS One 2017 Jul
27;12(7):e0181235.
[96] Ruan Q, Wang T, Kameswaran V, Wei Q, Johnson DS, Matschinsky F, et al. The
microRNA-21PDCD4 axis prevents type 1 diabetes by blocking pancreatic
b
cell death. Proc Natl Acad Sci U S A 2011 Jul 19;108(29):12030e5.
[97] Binger KJ, Neukam M, Tattikota SG, Qadri F, Puchkov D, Willmes DM, et al.
Atp6ap2 deletion causes extensive vacuolation that consumes the insulin
content of pancreatic
b
cells. Proc Natl Acad Sci USA 2019 Oct;116(40):
19983e8.
[98] Masini M, Bugliani M, Lupi R, del Guerra S, Boggi U, Filipponi F, et al.
Autophagy in human type 2 diabetes pancreatic beta cells. Diabetologia 2009
Jun 1;52(6):1083e6.
[99] Ji J, Petropavlovskaia M, Khatchadourian A, Patapas J, Makhlin J,
Rosenberg L, et al. Type 2 diabetes is associated with suppression of
autophagy and lipid accumulation in
b
-cells. J Cell Mol Med 2019 Apr;23(4):
2890e900.
[100] Cai T, Hirai H, Zhang G, Zhang M, Takahashi N, Kasai H, et al. Deletion of Ia-2
and/or Ia-2
b
in mice decreases insulin secretion by reducing the number of
dense core vesicles. Diabetologia 2011 Sep;54(9):2347e57.
[101] Torii S, Kubota C, Saito N, Kawano A, Hou N, Kobayashi M, et al. The
pseudophosphatase phogrin enables glucose-stimulated insulin signaling in
pancreatic
b
cells. J Biol Chem 2018 Apr 20;293(16):5920e33.
[102] Wollam J, Mahata S, Riopel M, Hernandez-Carretero A, Biswas A,
Bandyopadhyay GK, et al. Chromogranin A regulates vesicle storage and
mitochondrial dynamics to inuence insulin secretion. Cell Tissue Res 2017
Jun;368(3):487e501.
[103] Portela-Gomes GM, Gayen JR, Grimelius L, Stridsberg M, Mahata SK. The
importance of chromogranin A in the development and function of endocrine
pancreas. Regul Pept 2008 Nov 29;151(1e3):19e25.
[104] Paglialunga S, Simnett G, Robson H, Hoang M, Pillai R, Arkell AM, et al. The
Rab-GTPase activating protein, TBC1D1, is critical for maintaining normal
glucose homeostasis and
b
-cell mass. Appl Physiol Nutr Metabol 2017
Jun;42(6):647e55.
[105] Stermann T, Menzel F, Weidlich C, Jeruschke K, Weiss J, Altenhofen D, et al.
Deletion of the RabGAP TBC1D1 leads to enhanced insulin secretion and fatty
acid oxidation in islets from male mice. Endocrinology 2018 Apr 1;159(4):
1748e61.
[106] Laybutt DR, Preston AM, Åkerfeldt MC, Kench JG, Busch AK, Biankin AV,
et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type
2 diabetes. Diabetologia 2007 Apr 1;50(4):752e63.
[107] Engin F, Nguyen T, Yermalovich A, Hotamisligil GS. Aberrant islet unfolded
protein response in type 2 diabetes. Sci Rep 2014 Feb 11;4(1):4054.
[108] jiang Huang C, yu Lin C, Haataja L, Gurlo T, Butler AE, Rizza RA, et al.
High expression rates of human islet amyloid polypeptide induce endo-
plasmic reticulum stress mediated beta-cell apoptosis, a characteristic of
humans with type 2 but not type 1 diabetes. Diabetes 2007 Aug;56(8):
2016e27.
[109] Herbach N, Rathkolb B, Kemter E, Pichl L, Klaften M, de Angelis MH, et al.
Dominant-negative effects of a novel mutated Ins2 allele causes early-onset
diabetes and severe
b
-cell loss in Munich Ins2C95S mutant mice. Diabetes
2007 May 1;56(5):1268e76.
[110] Tiano JP, Mauvais-Jarvis F. Importance of oestrogen receptors to preserve
functional
b
-cell mass in diabetes. Nat Rev Endocrinol 2012 Jun;8(6):
342e51.
[111] Janson J, Soeller WC, Roche PC, Nelson RT, Torchia AJ, Kreutter DK,
et al. Spontaneous diabetes mellitus in transgenic mice expressing human
islet amyloid polypeptide. Proc Natl Acad Sci U S A 1996 Jul 9;93(14):
7283e8.
[112] Walker EM, Cha J, Tong X, Guo M, Liu JH, Yu S, et al. Sex-biased islet
b
cell
dysfunction is caused by the MODY MAFA S64F variant by inducing
premature aging and senescence in males. Cell Rep 2021 Oct 12;37(2):
109813.
[113] Mauvais-Jarvis F, Sobngwi E, Porcher R, Riveline JP, Kevorkian JP, Vaisse C,
et al. Ketosis-prone type 2 diabetes in patients of sub-saharan african origin:
clinical pathophysiology and natural history of
b
-cell dysfunction and insulin
resistance. Diabetes 2004 Mar 1;53(3):645e53.
Original Article
16 MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com
[114] Iacovazzo D, Flanagan SE, Walker E, Quezado R, de Sousa Barros FA,
Caswell R, et al. MAFA missense mutation causes familial insulinomatosis and
diabetes mellitus. Proc Natl Acad Sci U S A 2018 Jan 30;115(5):1027e32.
[115] Xu B, Allard C, Alvarez-Mercado AI, Fuselier T, Kim JH, Coons LA, et al.
Estrogens promote misfolded proinsulin degradation to protect insulin pro-
duction and delay diabetes. Cell Rep 2018 Jul 3;24(1):181.
[116] Zhou Z, Ribas V, Rajbhandari P, Drew BG, Moore TM, Fluitt AH, et al. Estrogen
receptor
a
protects pancreatic
b
-cells from apoptosis by preserving mito-
chondrial function and suppressing endoplasmic reticulum stress. J Biol
Chem 2018 Mar 30;293(13):4735.
[117] Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquié M,
Gauthier BR, et al. Pancreatic insulin content regulation by the estrogen
receptor ER alpha. PLoS One 2008 Apr 30;3(4):e2069.
[118] Cnop M, Ladriere L, Hekerman P, Ortis F, Cardozo AK, Dogusan Z, et al.
Selective inhibition of eukaryotic translation initiation factor 2
a
dephos-
phorylation potentiates fatty acid-induced endoplasmic reticulum stress and
causes pancreatic
b
-cell dysfunction and apoptosis. J Biol Chem 2007 Feb
9;282(6):3989e97.
[119] Oyadomari S, Koizumi A, Takeda K, Gotoh T, Akira S, Araki E, et al. Targeted
disruption of the Chop gene delays endoplasmic reticulum stressemediated
diabetes. J Clin Invest 2002 Feb 15;109(4):525e32.
[120] Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, et al. ER-stress-
induced transcriptional regulation increases protein synthesis leading to cell
death. Nat Cell Biol 2013 May;15(5):481e90.
[121] Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, et al. Diabetes
mellitus and exocrine pancreatic dysfunction in Perk/mice reveals a role
for translational control in secretory cell survival. Mol Cell 2001 Jun 1;7(6):
1153e63.
[122] Novoa I, Zhang Y, Zeng H, Jungreis R, Harding HP, Ron D. Stress-induced
gene expression requires programmed recovery from translational repres-
sion. EMBO J 2003 Mar 3;22(5):1180.
[123] Ma Y, Hendershot LM. Delineation of a negative feedback regulatory loop that
controls protein translation during endoplasmic reticulum stress. J Biol Chem
2003 Sep 12;278(37):34864e73.
[124] Panzhinskiy E, Skovsø S, Cen HH, Chu KY, MacDonald K, Soukhatcheva G,
et al. Eukaryotic translation initiation factor 2A protects pancreatic beta cells
during endoplasmic reticulum stress while rescuing translation inhibition.
bioRxiv 2021 Feb 17. 2021.02.17.431676.
[125] Cnop M, Toivonen S, Igoillo-Esteve M, Salpea P. Endoplasmic reticulum
stress and eIF2
a
phosphorylation: the Achilles heel of pancreatic
b
cells. Mol
Metabol 2017 Jul 12;6(9):1024e39.
[126] Sharma RB, ODonnell AC, Stamateris RE, Ha B, McCloskey KM, Reynolds PR,
et al. Insulin demand regulates
b
cell number via the unfolded protein
response. J Clin Invest 2015 Oct 1;125(10):3831e46.
[127] Sharma RB, Darko C, Alonso LC. Intersection of the ATF6 and XBP1
ER stress pathways in mouse islet cells. J Biol Chem 2020 Oct 9;295(41):
14164e77.
[128] Nasteska D, Fine NHF, Ashford FB, Cuozzo F, Viloria K, Smith G, et al.
PDX1LOW MAFALOW
b
-cells contribute to islet function and insulin release.
Nat Commun 2021 Jan 29;12(1):674.
[129] Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic
b
-cell dedifferentiation
as mechanism of diabetic
b
-cell failure. Cell 2012 Sep 14;150(6):1223.
[130] Hodeify R, Megyesi J, Tarcsafalvi A, Mustafa HI, Seng NSHL, Price PM.
Pathophysiology of Acute Kidney Injury: gender differences control the sus-
ceptibility to ER stress-induced acute kidney injury. Am J Physiol Ren Physiol
2013 Apr 1;304(7):F875.
[131] Manicardi V, Russo G, Napoli A, Torlone E, Li Volsi P, Giorda CB, et al.
Gender-disparities in adults with type 1 diabetes: more than a quality of care
issue. A cross-sectional observational study from the AMD annals initiative.
PLoS One 2016 Oct 3;11(10):e0162960.
MOLECULAR METABOLISM 69 (2023) 101678 Ó2023 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
www.molecularmetabolism.com 17
... Sexual dimorphism observed in glucose-stimulated [Ca 2+ ] c influx could be mediated by differences in Ca 2+ handling or differences in TALK-1 function. For example, under stress female human T2D islets maintain greater insulin secretion compared to males (Brownrigg et al., 2023). Although greater β-cell Kcnk16 expression could also be responsible for the more significant impairment in glucose intolerance observed in Kcnk16 L114P male mice, there most significantly (FDR <0.05) altered Gene Ontology (Molecular Function). ...
... for males, and average RPKM 7551±2952.70 for females) (Brownrigg et al., 2023). Whether sex differences exist in humans with TALK-1 L114P mutation remains to be determined as all affected individuals in both KCNK16-MODY (p. ...
Article
Full-text available
The gain-of-function mutation in the TALK-1 K ⁺ channel (p.L114P) is associated with maturity-onset diabetes of the young (MODY). TALK-1 is a key regulator of β-cell electrical activity and glucose-stimulated insulin secretion. The KCNK16 gene encoding TALK-1 is the most abundant and β-cell-restricted K ⁺ channel transcript. To investigate the impact of KCNK16 L114P on glucose homeostasis and confirm its association with MODY, a mouse model containing the Kcnk16 L114P mutation was generated. Heterozygous and homozygous Kcnk16 L114P mice exhibit increased neonatal lethality in the C57BL/6J and the CD-1 (ICR) genetic background, respectively. Lethality is likely a result of severe hyperglycemia observed in the homozygous Kcnk16 L114P neonates due to lack of glucose-stimulated insulin secretion and can be reduced with insulin treatment. Kcnk16 L114P increased whole-cell β-cell K ⁺ currents resulting in blunted glucose-stimulated Ca ²⁺ entry and loss of glucose-induced Ca ²⁺ oscillations. Thus, adult Kcnk16 L114P mice have reduced glucose-stimulated insulin secretion and plasma insulin levels, which significantly impairs glucose homeostasis. Taken together, this study shows that the MODY-associated Kcnk16 L114P mutation disrupts glucose homeostasis in adult mice resembling a MODY phenotype and causes neonatal lethality by inhibiting islet insulin secretion during development. These data suggest that TALK-1 is an islet-restricted target for the treatment for diabetes.
... males) is consistent with the recently evoked concept of βcell resilience in response to islet stress in female mice. 64 We did not observe major congruence in pathways identified by transcriptome and secretome analyses. This is expected as the secretome does not necessarily reflect the proteome signature but rather the pool of molecules packaged in the secretory granules ready to be released in the extracellular media under stimulatory conditions such as occurring experimentally in response to KCL 75 mM (Figure 1). ...
Article
Full-text available
Sensory neurons in the dorsal root ganglia (DRG) convey somatosensory and metabolic cues to the central nervous system and release substances from stimulated terminal endings in peripheral organs. Sex‐biased variations driven by the sex chromosome complement (XX and XY) have been implicated in the sensory–islet crosstalk. However, the molecular underpinnings of these male–female differences are not known. Here, we aim to characterize the molecular repertoire and the secretome profile of the lower thoracic spinal sensory neurons and to identify molecules with sex‐biased insulin sensing‐ and/or insulin secretion‐modulating activity that are encoded independently of circulating gonadal sex hormones. We used transcriptomics and proteomics to uncover differentially expressed genes and secreted molecules in lower thoracic T5‐12 DRG sensory neurons derived from sexually immature 3‐week‐old male and female C57BL/6J mice. Comparative transcriptome and proteome analyses revealed differential gene expression and protein secretion in DRG neurons in males and females. The transcriptome analysis identified, among others, higher insulin signaling/sensing capabilities in female DRG neurons; secretome screening uncovered several sex‐specific candidate molecules with potential regulatory functions in pancreatic β cells. Together, these data suggest a putative role of sensory interoception of insulin in the DRG–islet crosstalk with implications in sensory feedback loops in the regulation of β‐cell activity in a sex‐biased manner. Finally, we provide a valuable resource of molecular and secretory targets that can be leveraged for understanding insulin interoception and insulin secretion and inform the development of novel studies/approaches to fathom the role of the sensory–islet axis in the regulation of energy balance in males and females.
... Sex differences affect normal β-cell function and subsequent development of diabetes [126][127][128][129] . Therefore, we assessed sex differences across ages and their relationships to diabetes models. ...
Article
Full-text available
Although multiple pancreatic islet single-cell RNA-sequencing (scRNA-seq) datasets have been generated, a consensus on pancreatic cell states in development, homeostasis and diabetes as well as the value of preclinical animal models is missing. Here, we present an scRNA-seq cross-condition mouse islet atlas (MIA), a curated resource for interactive exploration and computational querying. We integrate over 300,000 cells from nine scRNA-seq datasets consisting of 56 samples, varying in age, sex and diabetes models, including an autoimmune type 1 diabetes model (NOD), a glucotoxicity/lipotoxicity type 2 diabetes model (db/db) and a chemical streptozotocin β-cell ablation model. The β-cell landscape of MIA reveals new cell states during disease progression and cross-publication differences between previously suggested marker genes. We show that β-cells in the streptozotocin model transcriptionally correlate with those in human type 2 diabetes and mouse db/db models, but are less similar to human type 1 diabetes and mouse NOD β-cells. We also report pathways that are shared between β-cells in immature, aged and diabetes models. MIA enables a comprehensive analysis of β-cell responses to different stressors, providing a roadmap for the understanding of β-cell plasticity, compensation and demise.
... In addition, transplantation of hESC-derived β-like cells into female recipient mice often yield faster in vivo maturation and insulin secretion properties than male recipient mice (58). Interactions between the MafA, MAFB, and nuclear sex hormone receptors (i.e., estrogen receptor β and androgen receptor) warrant further study, especially given the results described here and the sex-dependent differences in β cell gene expression found in normoglycemic mice (59) and by production of the S64F MAFA variant (60). ...
Article
Full-text available
Type 2 diabetes (T2D) is associated with compromised identity of insulin-producing pancreatic islet β cells, characterized by inappropriate production of other islet cell-enriched hormones. Here, we examined how hormone misexpression was influenced by the MAFA and MAFB transcription factors, closely related proteins that maintain islet cell function. Mice specifically lacking MafA in β cells demonstrated broad, population-wide changes in hormone gene expression with an overall gene signature closely resembling islet gastrin+ (Gast+) cells generated under conditions of chronic hyperglycemia and obesity. A human β cell line deficient in MAFB, but not one lacking MAFA, also produced a GAST+ gene expression pattern. In addition, GAST was detected in human T2D β cells with low levels of MAFB. Moreover, evidence is provided that human MAFB can directly repress GAST gene transcription. These results support a potentially novel, species-specific role for MafA and MAFB in maintaining adult mouse and human β cell identity, respectively. Here, we discuss the possibility that induction of Gast/GAST and other non-β cell hormones, by reduction in the levels of these transcription factors, represents a dysfunctional β cell signature.
Article
Full-text available
Diabetes constitutes a major public health problem, with dramatic consequences for patients. Both genetic and environmental factors were shown to contribute to the different forms of the disease. The monogenic forms, found both in humans and in animal models, specially help to decipher the role of key genes in the physiopathology of the disease. Here, we describe the phenotype of early diabetes in a colony of NOD mice, with spontaneous invalidation of Akt2, that we called HYP. The HYP mice were characterised by a strong and chronic hyperglycaemia, beginning around the age of one month, especially in male mice. The phenotype was not the consequence of the acceleration of the autoimmune response, inherent to the NOD background. Interestingly, in HYP mice, we observed hyperinsulinemia before hyperglycaemia occurred. We did not find any difference in the pancreas’ architecture of the NOD and HYP mice (islets’ size and staining for insulin and glucagon) but we detected a lower insulin content in the pancreas of HYP mice compared to NOD mice. These results give new insights about the role played by Akt2 in glucose homeostasis and argue for the ß cell failure being the primary event in the course of diabetes.
Chapter
Pancreas cell fate specification has been well studied in mice but is less documented in humans due to the lack of availability of pancreas from different developmental stages. Human pluripotent stem cells (hPSCs) can provide a model for human pancreas development that could be studied in vitro and in vivo as well as an unlimited source of cells that could be differentiated into glucose-responsive, insulin-producing cells and used to treat diabetes. The differentiation of pluripotent stem cell-derived pancreatic endoderm cells (PECs) into β cells that can ameliorate hyperglycemia in diabetic mouse models has been demonstrated in many studies, mostly in males. Given the current use of PECs in clinical trials, it is important to study how factors in the recipient may influence the differentiation of these PECs into endocrine cells. Sex, thyroid hormone levels, and site of implantation have been shown to impact the development of glucose-stimulated insulin secretion in differentiated PECs in mice. When these cells were implanted under the kidney capsule in female mice, they became glucose-responsive insulin-secreting cells faster than in male mice, and the formation of β cells was impaired by hypothyroidism. Additionally, when PECs were implanted in macroencapsulation devices, they not only differentiated into endocrine cells faster, but they were less likely to become ductal cells and form large cysts compared to PECs implanted under the kidney capsule or in the gonadal fat pad. Studies are underway to determine if these and other variables impact endocrine cell specification and function following PEC implant in humans.
Preprint
Full-text available
Caloric restriction (CR) extends organismal lifespan and health span by improving glucose homeostasis mechanisms. How CR affects organellar structure and function of pancreatic beta cells over the lifetime of the animal remains unknown. Here, we used single nucleus transcriptomics to show that CR increases the expression of genes for beta cell identity, protein processing, and organelle homeostasis. Gene regulatory network analysis link this transcriptional phenotype to transcription factors involved in beta cell identity (Mafa) and homeostasis (Atf6). Imaging metabolomics further demonstrates that CR beta cells are more energetically competent. In fact, high-resolution light and electron microscopy indicates that CR reduces beta cell mitophagy and increases mitochondria mass, increasing mitochondrial ATP generation. Finally, we show that long-term CR delays the onset of beta cell aging and senescence to promote longevity by reducing beta cell turnover. Therefore, CR could be a feasible approach to preserve compromised beta cells during aging and diabetes.
Preprint
A gain-of-function mutation in the TALK-1 K+ channel (p.L114P) associated with maturity-onset diabetes of the young (MODY) was recently reported in two distinct families. TALK-1 is a key regulator of β-cell electrical activity and glucose-stimulated insulin secretion (GSIS). KCNK16, the gene that encodes TALK-1, is the most abundant and β-cell– restricted K+ channel transcript; polymorphisms in the KCNK16 locus are also associated with an increased risk of type-2 diabetes. To investigate the impact of TALK-1-L114P on glucose homeostasis and confirm its association with MODY, a mouse model containing the Kcnk16 L114P mutation was generated. Heterozygous and homozygous Kcnk16 L114P mice exhibit increased neonatal lethality in the C57BL/6J and the mixed C57BL/6J:CD-1(ICR) genetic background, respectively. Lethality is likely a result of severe hyperglycemia observed in the homozygous Kcnk16 L114P neonates due to lack of GSIS and can be reduced with insulin treatment. TALK-1-L114P drastically increased whole-cell β-cell K+ currents resulting in blunted glucose-stimulated Ca2+ entry and loss of glucose-induced Ca2+ oscillations. Thus, adult Kcnk16 L114P mice have reduced GSIS and plasma insulin levels, which significantly impaired glucose homeostasis. Taken together, this study determined that the MODY-associated TALK-1-L114P mutation disrupts glucose homeostasis in adult mice resembling a MODY phenotype and causes neonatal lethality by altering islet hormone secretion during development. These data strongly suggest that TALK-1 is an islet-restricted target for the treatment of diabetes.
Article
Full-text available
Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic β-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent β-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the β-cell, identification of molecular markers that distinguish senescent from non-senescent β-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of β-cell senescence, highlighting insights from mouse models as well as studies on human islets and β-cells. We identify markers that have been used to detect β-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in β-cells, heterogeneity of senescent β-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.
Article
Full-text available
The importance of sexual dimorphism has been highlighted in recent years since the National Institutes of Health's mandate on considering sex as a biological variable. Although recent studies have taken strides to study both sexes side by side, investigations into the normal physiological differences between males and females are limited. In this study, we aimed to characterized sex-dependent differences in glucose metabolism and pancreatic β-cell physiology in normal conditions using C57BL/6J mice, the most common mouse strain used in metabolic studies. Here, we report that female mice have improved glucose and insulin tolerance associated with lower nonfasted blood glucose and insulin levels compared with male mice at 3 and 6 months of age. Both male and female animals show β-cell mass expansion from embryonic day 17.5 to adulthood, and no sex differences were observed at embryonic day 17.5, newborn, 1 month, or 3 months of age. However, 6-month-old males displayed increased β-cell mass in response to insulin resistance compared with littermate females. Molecularly, we uncovered sexual dimorphic alterations in the protein levels of nutrient sensing proteins O-GlcNAc transferase and mTOR, as well as differences in glucose-stimulus coupling mechanisms that may underlie the differences in sexually dimorphic β-cell physiology observed in C57BL/6J mice.
Article
Full-text available
Transcriptional and functional cellular specialization has been described for insulin-secreting β-cells of the endocrine pancreas. However, it is not clear whether β-cell heterogeneity is stable or reflects dynamic cellular states. We investigated the temporal kinetics of endogenous insulin gene activity using live cell imaging, with complementary experiments employing FACS and single cell RNA sequencing, in β-cells from Ins2GFP knock-in mice. In vivo staining and FACS analysis of islets from Ins2GFP mice confirmed that at a given moment, ∼25% of β-cells exhibited significantly higher activity at the conserved insulin gene Ins2. Live cell imaging captured Ins2 gene activity dynamics in single β-cells over time. Autocorrelation analysis revealing a subset of cells with oscillating behavior, with oscillation periods of 17 hours. Increased glucose concentrations stimulated more cells to oscillate and resulted in higher average Ins2 gene activity per cell. Single cell RNA sequencing determined that Ins2(GFP)HIGH β-cells were enriched for markers of β-cell maturity. Ins2(GFP)HIGH β-cells were also significantly less viable at all glucose concentrations and in the context of ER stress. Collectively, our results demonstrate that the heterogeneity of insulin production, observed in mouse and human β-cells, can be accounted for by dynamic states of insulin gene activity.
Article
Full-text available
Insulin receptor (Insr) protein is present at higher levels in pancreatic β-cells than in most other tissues, but the consequences of β-cell insulin resistance remain enigmatic. Here, we use an Ins1 cre knock-in allele to delete Insr specifically in β-cells of both female and male mice. We compare experimental mice to Ins1 cre -containing littermate controls at multiple ages and on multiple diets. RNA-seq of purified recombined β-cells reveals transcriptomic consequences of Insr loss, which differ between female and male mice. Action potential and calcium oscillation frequencies are increased in Insr knockout β-cells from female, but not male mice, whereas only male β Insr KO islets have reduced ATP-coupled oxygen consumption rate and reduced expression of genes involved in ATP synthesis. Female β Insr KO and β Insr HET mice exhibit elevated insulin release in ex vivo perifusion experiments, during hyperglycemic clamps, and following i.p . glucose challenge. Deletion of Insr does not alter β-cell area up to 9 months of age, nor does it impair hyperglycemia-induced proliferation. Based on our data, we adapt a mathematical model to include β-cell insulin resistance, which predicts that β-cell Insr knockout improves glucose tolerance depending on the degree of whole-body insulin resistance. Indeed, glucose tolerance is significantly improved in female β Insr KO and β Insr HET mice compared to controls at 9, 21 and 39 weeks, and also in insulin-sensitive 4-week old males. We observe no improved glucose tolerance in older male mice or in high fat diet-fed mice, corroborating the prediction that global insulin resistance obscures the effects of β-cell specific insulin resistance. The propensity for hyperinsulinemia is associated with mildly reduced fasting glucose and increased body weight. We further validate our main in vivo findings using an Ins1 -CreERT transgenic line and find that male mice have improved glucose tolerance 4 weeks after tamoxifen-mediated Insr deletion. Collectively, our data show that β-cell insulin resistance in the form of reduced β-cell Insr contributes to hyperinsulinemia in the context of glucose stimulation, thereby improving glucose homeostasis in otherwise insulin sensitive sex, dietary and age contexts.
Article
Full-text available
Objective Age is a risk factor for type 2 diabetes (T2D). We aimed to elucidate whether β cell glucose metabolism is altered with aging and contributes to T2D. Methods As a model of aging, we used senescence-accelerated mouse (SAM), C57BL/6J (B6), and ob/ob mice. As a diabetes model, we used db/db mice. Glucose responsiveness of insulin secretion and [U-¹³C]-glucose metabolic flux were examined in isolated islets. As molecular signatures of β cell identity, we analyzed expression of β-cell-specific genes in isolated islets and pancreatic sections. β cells defective in the malate-aspartate (MA) shuttle were previously generated from MIN6-K8 cells by knockout of Got1, a component of the shuttle. We analyzed Got1 KO β cells as a model of increased glycolysis. Results We identified hyperresponsiveness to glucose and compromised cellular identity as dysfunctional phenotypes shared in common between aged and diabetic mouse β cells. We also reveal a metabolic commonality between aged and diabetic β cells: hyperactive glycolysis through increased expression of Nmnat2, a cytosolic NAD-synthesizing enzyme. Got1 KO β cells showed increased glycolysis, β cell dysfunction, and impaired cellular identity, phenocopying aging and diabetes. Using Got1 KO β cells, we show that attenuation of glycolysis or Nmnat2 activity can restore β cell function and identity. Conclusion Our study demonstrates that hyperactive glycolysis is a metabolic signature of aged and diabetic β cells, which may underlie age-related β cell dysfunction and loss of cellular identity. We suggest Nmnat2 suppression as an approach to counteract age-related T2D.
Article
Full-text available
The Reactome Knowledgebase (https://reactome.org), an Elixir core resource, provides manually curated molecular details across a broad range of physiological and pathological biological processes in humans, including both hereditary and acquired disease processes. The processes are annotated as an ordered network of molecular transformations in a single consistent data model. Reactome thus functions both as a digital archive of manually curated human biological processes and as a tool for discovering functional relationships in data such as gene expression profiles or somatic mutation catalogs from tumor cells. Recent curation work has expanded our annotations of normal and disease-associated signaling processes and of the drugs that target them, in particular infections caused by the SARS-CoV-1 and SARS-CoV-2 coronaviruses and the host response to infection. New tools support better simultaneous analysis of high-throughput data from multiple sources and the placement of understudied ('dark') proteins from analyzed datasets in the context of Reactome's manually curated pathways.
Article
Full-text available
Type 2 diabetes mellitus (T2D) is a chronic age-related disorder characterized by hyperglycemia due to the failure of pancreatic beta cells to compensate for increased insulin demand. Despite decades of research, the pathogenic mechanisms underlying T2D remain poorly defined. Here, we use imaging mass cytometry (IMC) with a panel of 34 antibodies to simultaneously quantify markers of pancreatic exocrine, islet, and immune cells and stromal components. We analyze over 2 million cells from 16 pancreata obtained from donors with T2D and 13 pancreata from age-similar non-diabetic controls. In the T2D pancreata, we observe significant alterations in islet architecture, endocrine cell composition, and immune cell constituents. Thus, both HLA-DR-positive CD8 T cells and macrophages are enriched intra-islet in the T2D pancreas. These efforts demonstrate the utility of IMC for investigating complex events at the cellular level in order to provide insights into the pathophysiology of T2D.
Article
Full-text available
Type 2 diabetes (T2D) is a growing cause of poor health, psychosocial burden, and economic costs worldwide. The pancreatic β-cell is a cornerstone of metabolic physiology. Insulin deficiency leads to hyperglycemia, which was fatal before the availability of therapeutic insulins; even partial deficiency of insulin leads to diabetes in the context of insulin resistance. Comprising only an estimated 1 g or <1/500th of a percent of the human body mass, pancreatic β-cells of the islets of Langerhans are a vulnerable link in metabolism. Proinsulin production constitutes a major load on β-cell endoplasmic reticulum (ER), and decompensated ER stress is a cause of β-cell failure and loss in both type 1 diabetes (T1D) and T2D. The unfolded protein response (UPR), the principal ER stress response system, is critical for maintenance of β-cell health. Successful UPR guides expansion of ER protein folding capacity and increased β-cell number through survival pathways and cell replication. However, in some cases the ER stress response can cause collateral β-cell damage and may even contribute to diabetes pathogenesis. Here we review the known beneficial and harmful effects of UPR pathways in pancreatic β-cells. Improved understanding of this stress response tipping point may lead to approaches to maintain β-cell health and function.
Article
Full-text available
A heterozygous missense mutation of the islet β cell-enriched MAFA transcription factor (p.Ser64Phe [S64F]) is found in patients with adult-onset β cell dysfunction (diabetes or insulinomatosis), with men more prone to diabetes than women. This mutation engenders increased stability to the unstable MAFA protein. Here, we develop a S64F MafA mouse model to determine how β cell function is affected and find sex-dependent phenotypes. Heterozygous mutant males (MafAS64F/+) display impaired glucose tolerance, while females are slightly hypoglycemic with improved blood glucose clearance. Only MafAS64F/+ males show transiently higher MafA protein levels preceding glucose intolerance and sex-dependent changes to genes involved in Ca2+ signaling, DNA damage, aging, and senescence. MAFAS64F production in male human β cells also accelerate cellular senescence and increase senescence-associated secretory proteins compared to cells expressing MAFAWT. These results implicate a conserved mechanism of accelerated islet aging and senescence in promoting diabetes in MAFAS64F carriers in a sex-biased manner.
Article
In Drosophila, changes to dietary protein elicit different body size responses between the sexes. Whether these differential body size effects extend to other macronutrients remains unclear. Here, we show that lowering dietary sugar (0S diet) enhanced body size in male and female larvae. Despite an equivalent phenotypic effect between the sexes, we detected sex-specific changes to signaling pathways, transcription, and whole-body glycogen and protein. In males, the low sugar diet augmented insulin/insulin-like growth factor signaling pathway (IIS) activity by increasing insulin sensitivity, where increased IIS was required for male metabolic and body size responses in 0S. In females reared on low sugar, IIS activity and insulin sensitivity were unaffected, and IIS function did not fully account for metabolic and body size responses. Instead, we identified a female-biased requirement for the target of rapamycin pathway in regulating metabolic and body size responses. Together, our data suggest the mechanisms underlying the low sugar-induced increase in body size are not fully shared between the sexes, highlighting the importance of including males and females in larval studies even when similar phenotypic outcomes are observed.
Article
Male-female differences in many developmental mechanisms lead to the formation of two morphologically and physiologically distinct sexes. Although this is expected for traits with prominent differences between the sexes, such as the gonads, sex-specific processes also contribute to traits without obvious male-female differences, such as the intestine. Here, we review sex differences in developmental mechanisms that operate at several levels of biological complexity – molecular, cellular, organ and organismal – and discuss how these differences influence organ formation, function and whole-body physiology. Together, the examples we highlight show that one simple way to gain a more accurate and comprehensive understanding of animal development is to include both sexes.