Download as pdf or txt
Download as pdf or txt
You are on page 1of 2

Year in Review

ISLET BIOLOGY IN 2021 phosphorylation. Furthermore, based on


the disharmonic changes in β-​cell mRNA

New insights into β-cell failure, expression profiles and lack of alterations of
maturation and identity genes, the authors
suggested that β-​cells were not transdifferen-

regeneration and replacement tiating or dedifferentiating in T2DM. Previous


studies of pancreatic β-​cell dedi­fferentiation
in patients with T2DM have revealed con-
Mostafa Bakhti   and Heiko Lickert    tradictory results5,6, possibly due to differ-
ent patient characteristics, medication or
In 2021, several discoveries shed light on the pathomechanisms of β-​cell clinically determined T2DM subtypes. Data
failure during the initiation and progression of diabetes mellitus, and integration uncovered associations between
validated novel molecular targets for intervention. Moreover, the field plasma levels of ceramide, dihydroceramides
and ether-​linked phosphatidylcholines with
of stem-​cell-​derived replacements for β-​cells is rapidly advancing. These
HbA1c levels, suggesting their potential use
advances bring us closer to therapies to protect and/or regenerate β-​cell mass. as biomarkers4. Further analysis should apply
this multiple-​level data integration to different
A century after the discovery of insulin, no causal role of pancreatic exocrine dysfunction islet cell types and distinct β-​cell subpopula-
therapy can yet stop or reverse diabetes mel- in T1DM pathogenesis2. tions derived from different T2DM subtypes
litus progression1. The two major forms of Chiou and colleagues3 then took a sim- to identify the disease proteins and pathways.
diabetes mellitus include type 1 diabetes mel- ilar approach to investigate the genetic Coming from a completely different
litus (T1DM) and type 2 diabetes mellitus basis of T2DM risk. Single-​cell ATAC-​seq angle, a screen for novel endocrine regulators
(T2DM), which cause loss and dysfunction analysis of healthy human islets provided during pancreas development identified
of pancreatic β-​cells, respectively. Thus, it is lineage-​specific and state-​specific accessible a novel regulator of insulin/insulin-​like
crucial to uncover the molecular mechanisms chromatin profiles, indicating the existence of growth factor (IGF) signalling7 called insu-
of β-​cell pathophysiology to design therapies epigenomic heterogeneity in endocrine cells3. lin inhibitory receptor (inceptor). Whether
for protecting or restoring functional β-​cell The mapping of cis-​regulatory programmes insulin-​producing β-​cells experience auto-
mass. In 2021, several studies investigated onto SNP-​based GWAS of T2DM identified crine insulin signalling at the source of
the pathological changes to islets in T1DM enrichment of many T2DM risk variants, the ligand and how they are shielded from
and T2DM, identified a novel drug target including rs231361 at the KCNQ1 locus constitutive pathway activation are points
and established a new islet transplantation (Fig. 1). Data analysis predicted state-​specific of controversy in the field of β-​c ell biol-
strategy. These advances open new ave- effects of rs231361 on β-​c ell chromatin ogy. The identification and characteriza-
nues for β-​cell protection, regeneration and accessibility. Experiments with gene-​edited tion of inceptor (Fig. 1) shed some new light
replacement. human embryonic stem-​cell-​derived β-​like onto these old questions 7. Whole-​ b ody
The cause and mechanisms of T1DM are cells revealed that rs231361 regulates insulin knockout of inceptor resulted in increased
poorly understood. Several genome-​wide synthesis, further highlighting the functional β-​c ell insulin signall­ing  and β-​c ell mass,
association studies (GWAS) have identified impact of noncoding genetic risk variants
genetic risk variations linked with T1DM in T2DM3. Future studies should function-
Key advances
susceptibility, largely at noncoding genomic ally validate more of the identified risk var-
regions. Nevertheless, the causality of these iants to detect more T1DM-​associated and • Single-​cell epigenomics and genome-​wide
variants to the onset and progression of T2DM-​associated disease genes and potential association studies (GWAS) of type 1
T1DM remains unclear. In an elegant study therapeutic targets. diabetes mellitus (T1DM) detected
by Chiou et al.2, GWAS data were combined Profiling of islets from people with diabe- enrichment of risk variants in cis-​regulatory
elements, implicating the contribution of
with single-​c ell epigenomics to map the tes mellitus is hampered by possible molecular
pancreatic exocrine dysfunction to the
genetic basis of T1DM. The investigators used changes during isolation of the samples from development of T1DM2.
single-​nucleus assay for transposase-​accessible deceased donors and lack of natural history
• Combining single-​cell epigenomics and
chromatin using sequencing (ATAC-​seq) to of the disease development. To overcome this GWAS of type 2 diabetes mellitus (T2DM)
generate an epigenomic map of candidate hurdle, Wigger et al.4 snap-​froze pancreatic identified a risk variant at the KCNQ1 locus,
cis-​regulatory elements (cCREs) in peripheral tissue from metabolically profiled living indi- which impacts insulin synthesis3.
blood samples and pancreas biopsy samples viduals undergoing pancreatectomies and • Multi-​omics analysis of samples from deeply
from individuals without any form of diabe- isolated islets using laser-​capture microdis- metabolically profiled individuals
tes mellitus2. By performing a large single- section. These individuals represented a wide uncovered molecular changes associated
nucleotide-​ p olymorphism (SNP)-​ b ased range of glycaemia conditions, including nor- with β-​cell pathophysiology in T2DM and
GWAS of T1DM and integrating it with the moglycaemia, impaired glucose tolerance and identified novel lipid biomarkers4.
epigenome, they discovered enrichment of overt T2DM4. The researchers performed bulk • The unexpected discovery of an insulin
many T1DM risk variants in cell-​type-​specific mRNA sequencing and proteomics of isolated inhibitory receptor (inceptor) provided
cCREs (Fig. 1). These risk variants included ref- islets and conducted plasma lipidomics (Fig. 1). insights into how insulin signalling and the
erence SNP (rs)7795896, which they mapped The transcriptomic analysis revealed progres- insulin receptor are regulated in β-​cells7.
to a ductal-​cell-​specific cCRE. Functional sive alterations in gene expression patterns in • Ready-​made microvessels improved the
analysis revealed that rs7795896 suppresses impaired glucose tolerance and islets from survival and function of transplanted stem-​cell-
expression of cystic fibrosis transmembrane people with T2DM, including in genes asso- derived islets and primary human islets into
mice, enhancing T1DM remission9.
conductance regulator, uncovering a possible ciated with insulin secretion and oxidative


NATuRe RevieWS | ENdocriNology volume 18 | FEBRUARY 2022 | 79

0123456789();:
Year in Review

Single-cell
epigenomics

T1DM and
T2DM GWAS
Pancreatic Peripheral blood
tissues mononuclear cells
SC-islets Ready-made Data integration and
or primary microvessels functional validation to
human islets identify causal risk variants
Postmortem
tissue samples

Design drugs: Major regulators of


Co-transplantation • Small molecules β-cell development
Protection and • Targeted delivery and function, such
• Improved engraftment • Antibodies as inceptor
regeneration
• Increased β-cell survival,
maturation and function
• Enhanced diabetes • Transcriptomics
mellitus remission Samples from
live donors • Proteomics
• Lipidomics

Islets from healthy


donors and people
with T2DM

Fig. 1 | Novel approaches and targets for β-cell protection, regener- diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Identifying
ation and replacement. Combining single-​c ell epigenomics of inceptor provides a new target for enhancing insulin signalling to
human pancreatic cells with genome-​wide association study (GWAS) data restore β-​cell mass. Co-​transplanting stem-​cell-​derived islets (SC-​islets)
and multi-​omics analyses of pancreatic biopsy samples from living or primary human islets with ready-​m ade microvessels enhances
individuals revealed novel genes linked with β-​cell failure in type 1 engraftment.

✉e-​mail: heiko.lickert@helmholtz-​muenchen.de
causing hyperinsulinaemia and hypogly- restore normoglycaemia in various mouse
caemia in mice. Inceptor physically inter- models of diabetes mellitus. Importantly, https://doi.org/10.1038/s41574-021-00611-0
acts with insulin and IGF1 receptors and microvessels also enhanced the survival and 1. Sims, E. K., Carr, A. L. J., Oram, R. A., DiMeglio, L. A.
facilitates clathrin-​mediated endocytosis function of grafted primary human islets9. & Evans-​Molina, C. 100 years of insulin: celebrating
the past, present and future of diabetes therapy.
of the activated insulin and IGF receptor Despite these exciting findings, more refine- Nat. Med. 27, 1154–1164 (2021).
complexes to desensitize insulin and IGF ments are still required. For example, combin- 2. Chiou, J. et al. Interpreting type 1 diabetes risk with
genetics and single-​cell epigenomics. Nature 594,
signalling in β-​cells. Importantly, mice lack- ing an encapsulation device10 with microvessels 398–402 (2021).
ing inceptor specifically in β-​cells exhibited could deliver functional SC-​islets and primary 3. Chiou, J. et al. Single-​cell chromatin accessibility
increased β-​cell proliferation and improved human islets more efficiently through reducing identifies pancreatic islet cell type- and state-​specific
regulatory programs of diabetes risk. Nat. Genet. 53,
glucose tolerance 7. As total insulin and graft rejection by the host immune system. 455–466 (2021).
IGF1 resistance in β-​cells leads to diabetes These studies have important implications 4. Wigger, L. et al. Multi-​omics profiling of living human
pancreatic islet donors reveals heterogeneous beta
mellitus8, ways of sensitizing β-​cells to insu- for deciphering the pathomechanisms of cell trajectories towards type 2 diabetes. Nat. Metab.
lin and IGF signalling might prevent β-​cell β-​cell failure and developing new approaches 3, 1017–1031 (2021).
5. Butler, A. E. et al. β-​cell deficit in obese type 2
loss and dysfunction in diabetes mellitus. to regenerate or replace them. Continuing diabetes, a minor role of β-​cell dedifferentiation
Another strategy to restore β-​cell mass is advancements in single-​cell multi-​omics and and degranulation. J. Clin. Endocrinol. Metab. 101,
523–532 (2016).
replacing lost β-​cells by transplanting cadaveric functional validations will provide deeper 6. Cinti, F. et al. Evidence of β-​cell dedifferentiation in
human islets or in vitro-​differentiated pluri- insights into β-​cell dysfunction and find novel human type 2 diabetes. J. Clin. Endocrinol. Metab.
101, 1044–1054 (2016).
potent stem-​cell-​derived islet-​like clusters targets for β-​cell protection and/or regener- 7. Ansarullah et al. Inceptor counteracts insulin signalling
(SC-​islets). However, this approach requires ation. These advancements — together with in β-​cells to control glycaemia. Nature 590, 326–331
(2021).
optimization on graft delivery, graft–host improvements in regenerative medicine — 8. Ueki, K. et al. Total insulin and IGF-​I resistance in
interactions and graft survival, safety and will improve the quality of life for people with pancreatic β cells causes overt diabetes. Nat. Genet.
38, 583–588 (2006).
function (Fig. 1). In a study by Aghazadeh et al.9, diabetes mellitus. 9. Aghazadeh, Y. et al. Microvessels support engraftment
adipose-​tissue-​derived ready-​made microves- Mostafa Bakhti   1,2 and Heiko Lickert   1,2,3 ✉
and functionality of human islets and hESC-​derived
pancreatic progenitors in diabetes models. Cell Stem
sels were co-​transplanted with human SC-​islets, Institute of Diabetes and Regeneration Research,
1 Cell 28, 1936–1949.e8 (2021).
increasing graft survival, expansion and β-​cell Helmholtz Zentrum München, Neuherberg, Germany.
10. Wang, X. et al. A nanofibrous encapsulation device
for safe delivery of insulin-​producing cells to treat
maturation (Fig. 1). These results were attributed German Center for Diabetes Research (DZD),
2 type 1 diabetes. Sci. Transl. Med. 13, eabb4601
to the efficient integration of the microves- Neuherberg, Germany. (2021).

sels with the host vasculature system, which 3


Technische Universität München, School of Medicine, Competing interests
improved function of the grafted SC-​islets to Munich, Germany. The authors declare no competing interests.

80 | FEBRUARY 2022 | volume 18 www.nature.com/nrendo

0123456789();:

You might also like