You are on page 1of 8

Acta Biochim Biophys Sin, 2020, 00(00), 1–8

doi: 10.1093/abbs/gmaa047
Review

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
Review

Recent advances and potential applications of


human pluripotent stem cell-derived pancreatic
β cells
Ziyu Zhou, Xiaojie Ma, and Saiyong Zhu*
MOE Laboratory of Biosystems Homeostasis and Protection, Life Sciences Institute, Zhejiang University,
Hangzhou 310058, China
∗ Correspondence address. Tel/Fax: +86-571-88981797; E-mail: saiyong@zju.edu.cn
Received 26 December 2019; Editorial Decision 23 February 2020

Abstract
Diabetes mellitus is characterized by chronic high blood glucose levels resulted from deficiency
and/or dysfunction of insulin-producing pancreatic β cells. Generation of large amounts of func-
tional pancreatic β cells is critical for the study of pancreatic biology and treatment of diabetes.
Recent advances in directed differentiation of pancreatic β-like cells from human pluripotent stem
cells (hPSCs) can provide patient-specific and disease-relevant target cells. With the improved
differentiation protocols, it is now possible to generate large amounts of functional human
pancreatic β-like cells that can response to high level of glucose both in vitro and in vivo. Combined
with precise genomic editing, biomedical engineering, high throughput profiling, bioinformatics,
and high throughput genetic and chemical screening, these hPSC-derived pancreatic β-like cells
will hold great potentials in disease modeling, drug discovery, and cell-based therapies. In this
review, we summarize the recent progress in human pancreatic β-like cells derived from hPSCs
and discuss their potential applications.

Key words: pancreatic β cells, human pluripotent stem cells, disease modeling, drug discovery, cell-based therapy

Introduction Human pluripotent stem cells (hPSCs), including human embry-


According to the International Diabetes Federation, diabetes mellitus onic stem cells (hESCs) and human-induced pluripotent stem cells
represents a global health epidemic and affects more than 300 mil- (hiPSCs), are very promising sources to provide unlimited supplies
lion people worldwide. As an inevitable consequence, high blood of functional pancreatic β-like cells. hPSCs have infinite self-renewal
glucose can lead to many serious life-threatening diseases, including capacity and can be differentiated into any specific cell types of the
amputation, blindness, neuropathy, stroke, cardiovascular diseases, human body. Very encouragingly, many efforts have already been
and kidney diseases. While the use of insulin has become a widely made to generate an unlimited supply of functional pancreatic β-like
used therapeutic intervention, other promising treatment options cells from hPSCs, and in combination with an encapsulation device
have also been actively investigated at academic and pre-clinic stages. or patient-specific autologous iPSCs [2]. While significant progresses
Notably, the Edmonton protocol for islet transplantation has already have been made with these innovative strategies, novel and cost-
shown encouraging results and will undoubtedly hold great promises effective approaches are undoubtedly required for more successful
in treating or even curing diabetes, but there remain many hurdles, treatments and the ultimate cures for diabetes.
including very limited supplies of healthy human islets and the issues In this review, we will summarize recent advances on human
of life-long immunosuppression [1]. Therefore, we need to develop pancreatic β-like cell differentiation from hPSCs and the potential
novel approaches to prepare patient-specific functional pancreatic β applications of these hPSC-derived pancreatic β-like cells in disease
cells for both basic studies and clinical applications. modeling, drug discovery, and cell-based therapies.

© The Author(s) 2020. Published by Oxford University Press on behalf of the Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese
Academy of Sciences. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com 1
2 Pancreatic β cells from hPSCs

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
Figure 1. Generation of hPSC-derived functional pancreatic β-like cells (A) The human pancreatic differentiation in vitro mimics human pancreas development
in vivo. (B) Timeline of four key directed human pancreatic differentiation protocols. In 2008, Kroon et al. [3] reported a five-stage protocol to obtain PPs from
hPSCs. These PP cells could mature in vivo. In 2014, Rezania et al. [10] demonstrated a seven-stage protocol to generate insulin-secreting pancreatic β-like
cells from hPSCs. Meanwhile, Pagliuca et al. [11] developed a scalable 3D differentiation protocol to generate SC-β cells from hPSCs. In 2019, Nair et al. [17]
optimized their protocols to produce more mature pancreatic β-like cells. They isolated INS-GFP-positive cells and reaggregated them into islet-sized eBCs.
Significantly, these eBCs had better glucose-stimulated insulin-secreting functions both in vitro and in vivo. Growth factors and small molecules: Act A, Activin A;
Wnt, Wnt3a; RA, retinoic acid; Cyc, KAAD-cyclopamine; Vc, vitamin C; TPB, (2S,5S)-(E,E)-8-(5-(4-(trifluoromethyl) phenyl)-2,4 pentadienoylamino) benzolactam;
SANT, Sonic hedgehog antagonist; LDN, LDN193189; ALK5iII, ALK5 inhibitor II; T3, triiodothyronine; N-Cys, N-acetyl cysteine; CHIR, CHIR99021; PDBu, Phorbol
12,13-dibutyrate; XXI, γ-secretase inhibitor XX; TTNPB, (E)-4-(2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propen-1-yl) benzoic acid.

et al. [3] further optimized their pancreatic differentiation protocol


Generation of Functional Pancreatic β-like Cells
and demonstrated that functional endocrine cells could be generated
from hPSCs in vivo after transplanting PPs. Several months after implantation,
Over the past two decades, tremendous efforts have been made these hESC-derived pancreatic β-like could reverse hyperglycemia
to differentiate hPSCs into insulin-secreting pancreatic β-like cells. induced by streptozotocin. Besides growth factors, small molecules
Briefly, stepwise differentiation protocols have been developed to also play critical roles in human pancreatic differentiation [7]. For
guide the directed differentiation of hPSCs into definitive endoderm example, in 2009, Chen et al. [8] found that PKC activator ILV
(DE), primitive gut tube, posterior foregut, and pancreatic progen- could increase the percentage of hESC-derived PDX1-positive cells
itors (PPs). After transplantation into immunodeficient mice, these through a large-scale chemical screening. In 2012, Rezania et al.
PP cells can further mature into functional pancreatic β-like cells [9] optimized the 2D differentiation system and demonstrated that
in vivo [3]. Subsequently, with improved differentiation protocols, the combination of Alk5 inhibitor with PKC activator TPB and
functional pancreatic β-like cells (PB) can be derived from hPSCs BMP pathway inhibitor noggin at stage 4 could increase the expres-
under in vitro cell culture conditions (Fig. 1). sion of NKX6.1. In 2014, there were several breakthroughs of
In 2001, Assady et al. [4] first demonstrated that pancreatic β-like pancreatic differentiation protocols. Rezania et al. [10] developed
cells could be randomly differentiated from hESCs using the method an improved seven-stage differentiation protocol, which can finally
of embryonic bodies (EBs), albeit with very limited efficiency. In produce ∼50% insulin-positive cells. At first, they added vitamin C at
2005, D’Amour et al. [5] identified that SOX17 and FOXA2 double- stages 2 and 3 to suppress the expression of NGN3 and used thyroid
positive DE could be efficiently induced from hESCs by high concen- hormone (T3) at stage 5 to enhance the co-expression of NKX6.1
tration of activin A and low concentration of serum. The next year, and insulin. Next, through a small-scale chemical screening, they
the same group reported a five-stage protocol for the generation of identified that R428, a tyrosine kinase receptor AXL inhibitor, could
insulin-producing endocrine cells from hESCs [6]. In 2008, Kroon induce MAFA expression. Finally, they demonstrated that air-liquid
Pancreatic β cells from hPSCs 3

interface culture from stage 5 to 7 could promote the generation of human early development. High-throughput sequencing approaches
functional pancreatic β-like cells in vitro. Although these stage-7 cells have helped researchers to have a better understanding of human
were not identical to adult islet β cells, they could secrete insulin pancreatic development and differentiation. In 2017, Ameri et al. [21]
and ameliorate diabetes within 40 days. Pagliuca et al. [11] reported identified a specific PP surface marker glycoprotein 2 (GP2) using

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
a scalable 3D differentiation protocol to generate large amounts a comparative gene expression analysis. The enrichment of GP2-
of hPSC-derived pancreatic β-like cells (SC-β cells). After testing positive PPs could improve pancreatic β-like cell differentiation effi-
more than 150 combinations involving over 70 compounds, they ciency. Recently, single-cell RNA-seq (scRNA-seq) has become a pro-
used a combination of 11 small molecules to obtain functional SC-β mising technique to identify and characterize cell types within organs
cells. These SC-β cells contained insulin granules, responded to the and tissues. Using scRNA-seq, Veres et al. [22] characterized different
change of glucose level in vitro, and reversed hyperglycemia in vivo. cell types emerging during human pancreatic β-like cell differentia-
However, the authors also noted that these SC-β cells showed less tion. After bioinformatic analysis, they found that CD49a could be
functions compared with bona fide β cells, and more improvements used as a specific surface marker of pancreatic β-like cells derived from
were still needed. In 2015, Russ et al. [12] provided a fast and hPSCs. Later, they magnetically sorted CD49a-positive pancreatic β-
scalable approach to produce functional human pancreatic β-like like cells with a purity up to 80%. Therefore, this work provided a
cells. They demonstrated that the depletion of cyclopamine and novel perspective to increase the percentage of pancreatic β-like cells.
Noggin at stages 3 and 4 could increase the percentage of PDX1 and Interestingly, Alvarez-Dominguez et al. [23] analyzed the epigenome
NKX6.1 double-positive PPs. Interestingly, they found that precise dynamics of human pancreatic differentiation and demonstrated
temporal activation of endocrine differentiation was critical for the that circadian rhythms could be used to stimulate pancreatic islet
final production of functional pancreatic β-like cells. In addition, maturation. High-throughput profiling approaches will facilitate
these hPSC-derived pancreatic β-like cells could response to high researchers to have a better understanding of human pancreatic
level of glucose in vitro and protect mice from diabetes in vivo differentiation and development on multiple ‘omic’ levels, including
after transplantation. However, compared with adult pancreatic β genomic, epigenomic, transcriptomic, proteomic, and metabolomic.
cells, these hPSC-derived pancreatic β-like cells still performed less The combination of CRISPR genome editing and differentiation
functional and needed further improvements. system provided another powerful platform to study human pan-
Recently, scientists worldwide have made their efforts on opti- creatic differentiation and development. CRISPR genome editing
mizing the pancreatic differentiation strategies from various aspects approaches, including CRISPR-Cas9 [24] and CRISPR-Cpf1 [25],
using 3D culture system, biomimetic materials and biomedical engi- have many applications in human stem cell biology [24–27]. For
neering [13–17]. Interestingly, by mimicking early embryonic devel- example, Wei et al. [26] performed a genomic CRISPR knockout
opment, 3D culture system can promote the proliferation of PPs screen and identified that VDR is important for pancreatic β cell
and their further differentiation into pancreatic β-like cells [18–20]. maintenance and function. Later, they discovered that vitamin D
In 2017, Wang et al. [13] demonstrated that a specific biomimetic could switch BAF complexes to protect pancreatic β cells both in vitro
3D scaffold could help the generation of pancreatic endoderm and and in vivo. In another study, Li et al. [27] carried out genome-scale
endocrine cells from hPSCs. In 2018, Mamidi et al. [14] found CRISPR screens and identified Jun N-terminal kinase-JUN family
that the extracellular matrix could affect the fate choices of bi- genes as DE barriers. Similar strategies can be applied to identify
potential PPs through F-actin-YAP1-Notch mechano-signaling. In critical barriers or enhancers of human pancreatic differentiation at
2019, Youngblood et al. [15] used a microporous biomaterial to various stages and improve pancreatic β-cell functions.
guide PPs to form defined-sized clusters and promote pancreatic β- In summary, many significant advances have recently been made
like cell maturation. Very recently, Velazco-Cruz et al. [16] optimized in pancreatic differentiation protocols. As shown in Fig. 1, the proto-
the directed differentiation protocol by resizing the pancreatic β- cols for the derivation of PPs are becoming more consistent, efficient,
like cell clusters. Significantly, Nair et al. [17] developed cell culture and reproducible. Additionally, insulin-producing pancreatic β-like
conditions to closely mimic events occurring during pancreatic islet cells can now be differentiated from these PP cells with various
organogenesis and β cell maturation. They isolated INS-GFP-positive efficiencies. However, even with the most recent protocols, these
cells by FACS and then reaggregated these pancreatic β-like cells hPSC-derived pancreatic β-like cells are still less mature than primary
to form islet-sized enriched β-clusters (eBCs). These eBCs could islet β cells. One solution is to put more efforts on further optimizing
display many physiological properties analogous to primary human these in vitro culture systems to maintain the terminally differentiated
pancreatic β cells, including robust dynamic insulin secretion in pancreatic β cells. Another approach is to promote maturation
vitro, improved mitochondrial energization, and much better in vivo through transplantation and taking advantage of in vivo niches.
functions. Moreover, incorporating pancreatic niche cells, including As we have discussed earlier, better understanding of pancreatic
endothelial and mesenchymal cells, will also be beneficial. In the development of model organisms and human beings are the major
future, various tissue engineering strategies can be applied to the gen- keys to generate mature and functional target cells. In the future,
eration of more mature and functional pancreatic cells and tissues. the rapid advances of high-throughput profiling, high-throughput
In principle, current pancreatic differentiation strategies largely genome-wide genetic screening, high-throughput chemical screening,
mimic pancreatic development during embryogenesis. In the past few and biomedical engineering will undoubtedly provide novel infor-
decades, a few model organisms, such as zebrafish, frog, and mouse, mation and perspectives to human pancreas development and help
have been very effectively used to describe the normal development develop better pancreatic differentiation approaches.
of the pancreas. These studies have identified key genes and signaling
pathways that can guide projects aimed at turning hPSCs into pan-
Applications of hPSC-derived Pancreatic β-like
creatic β cells. It is necessary to understand how human pancreatic β
Cells in Disease Modeling
cells are normally made and use that information to make functional
human pancreatic β-like cells that can be used to understand and treat During the past decades, the advances of genome-wide association
diabetes. However, it is technically and ethically challenging to study studies (GWASs), hiPSC technique, directed pancreatic differentia-
4 Pancreatic β cells from hPSCs

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
Figure 2. Applications of hPSC-derived pancreatic β-like cells in disease modeling Unlimited number of functional pancreatic β-like cells can be generated by
directed differentiation from hPSCs. Combined with CRISPR genome editing and high-throughput profiling, these pancreatic β-like cells can be used for disease
modeling and to identify the underlying molecular mechanisms of the progression of diabetes.

combined with CRISPR genome editing [28]. In particular, there are


at least 11 described maturity onset diabetes of the young (MODY)
resulted from genetic mutations at specific genes, including MODY1
(HNF4A), MODY2 (GCK), MODY3 (HNF1A), MODY4 (PDX1),
MODY5 (HNF1B), MODY6 (NEUROD1), MODY7 (KLF11),
MODY8 (CEL), MODY9 (PAX4), MODY10 (INS), and MODY11
(BLK). Besides MODYs, neonatal diabetes is another common
form of monogenic diabetes caused by mutations in genes, such
as KCNJ11 and ABCC8. Recently, Flanagan et al. [29] identified
NKX2.2 and MNX1 as etiological genes for neonatal diabetes
and confirmed their critical roles in the development of human
pancreas. In addition, mutations in mitochondrial DNA (mtDNA)
can cause mitochondrial diabetes. The most common one linked
to mitochondrial diabetes is 3243 A>G in the MT-TL1 gene. As
carrying out mtDNA genome editing remains very difficult, patient-
specific hiPSC technologies can provide feasible ways to study this
type of diabetes.
With the advent of CRISPR-Cas system, many laboratories have
already carried out hPSC-based modeling of diabetes (Fig. 2). In
2016, Zhu et al. [30] studied eight key genes in pancreatic develop-
ment by combining CRISPR-Cas9 and directed pancreatic differenti-
ation. They found that PDX1 had haploinsufficient effect in human
pancreatic differentiation, and RFX6 could regulate the number of
PPs and endocrine differentiation. In addition, they demonstrated
that NGN3 plays different roles in humans and in mice. The loss
of NGN3 led to the absence of pancreatic β cells in mice, but a small
number of pancreatic β-like cells formed from NGN3-knockout
hPSCs. In the same year, Zeng et al. [31] combined GWAS and
Figure 3. Applications of hPSC-derived pancreatic β-like cells in drug
directed differentiation system to study diabetes-related mutations
discovery Pancreatic β-like cells, which are derived from hiPSCs or
genome-edited hPSCs, carry many hallmarks of patient-specific genetic and
in CDKAL1, KCNQ1, and KCNJ11. They found that biallelic
epigenetic states and are suitable for both target-focused and phenotypic mutations in these three genes did not influence the generation of
large-scale chemical screening. After further in vitro assays and animal-based pancreatic β-like cells from hPSCs but affected glucose-stimulated
evaluations, these drug candidates will potentially provide new options for insulin secretion. In 2017, Shi et al. [32] reported that the deletion
personalized therapy. of GATA6 impaired the differentiation of DEs from hPSCs. Inter-
estingly, GATA6 haploinsufficiency led to less PPs and β-like cells.
In addition, the haploinsufficiency of GATA6 was also affected by
tion protocols, and CRISPR genome editing systems have opened GATA4 dosage. This study shed the new light on disease modeling
new avenues for modeling and studying diabetes. Diabetes is a from the individual genetic defects to multigenetic modifications. In
multifaceted and multifactorial disease. Many genetic mutations 2018, Amin et al. [33] investigated the role of GLIS3, which is asso-
can cause diabetes and can be modeled and investigated using hPSCs ciated with T1D, T2D, and neonatal diabetes, in pancreatic differen-
Pancreatic β cells from hPSCs 5

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
Figure 4. Potential applications of hPSC-derived pancreatic β-like cells in cell-based therapy There are many critical issues need to be addressed, including
immune rejection, safety, cost, and long-term functions. Several promising strategies, including surface modification, encapsulation, and precise genome
editing, are under active investigation. Optimistically, these efforts will eventually lead to significant advances in cell-based therapy.

tiation. They found that GLIS3 deletion impaired the differentiation loci and then identify candidate drugs using high-throughput chemi-
of PDX1 and NKX6.1 double-positive PPs from hPSCs. Mechanisti- cal screening. For example, Zeng et al. [31] found that the CDKAL1
cally, they figured out that the knockout of GLIS3 led to pancreatic mutations could cause impaired glucose-stimulated insulin secretion
β-like cell death by activating the TGF-β pathway. In another study, both in vitro and in vivo. These CDKAL1 knockout pancreatic β-like
Zhou et al. [34] reported that a commonly-used pesticide propargite cells were hypersensitive to glucotoxicity and lipotoxicity. In order
could induce β-like cell death. The loss of GSTT1, which increases to protect CDKAL1−/− pancreatic β-like cells from glycolipid tox-
the risk of diabetes, made pancreatic β-like cells hypersensitive to icity, they performed a high-content compound screening and then
propargite-induced cell death. Their study demonstrated the role of identified a candidate drug T5224 that could rescue CDKAL1−/− -
gene–environment interaction on diabetes. In 2019, Lee et al. [35] specific defects by inhibiting the FOS/JUN pathway. In another
demonstrated that FOXA2 was required in the formation of PPs. study from the same research group, Amin et al. [33] explored
The loss of FOXA2 would reduce the number of PPs during the the roles of GLIS3 gene in human pancreatic development. Using
directed differentiation process and impair the recruitment of GATA6 improved differentiation protocols, they found that GLIS3−/− hESCs
to pancreatic enhancers. The coalescence of disease modeling and showed impaired pancreatic differentiation with significant death
high-throughput profiling provides novel information to understand of pancreatic β-like cells. With a high-content chemical screening,
pancreatic development. Significantly, this phenotype was not seen they found that a drug candidate, galunisertib, could effectively
in Foxa2 conditional knockout mice. Recently, Cardenas-Diaz et inhibit the inappropriate upregulation of the TGF-β pathway and
al. [36] reported that HNF1A could inhibit α cell gene expression, rescue apoptosis of pancreatic β-like cells induced by GLIS3 deletion.
regulate cellular metabolism, and maintain pancreatic endocrine cell Therefore, the application of the directed pancreatic differentiation
function. Then, they found that HNF1A could target a human- system provides unlimited cell sources for drug screening with the
specific long non-coding RNA, LINKA, which could regulate normal context of human pancreas.
mitochondrial respiration. Using similar approaches, other diabetes- Types 1 and 2 diabetes are both resulted from a deficiency
related mutations and more complicated subtypes can be modeled. of functional pancreatic β cells. Although pancreatic β cells were
In the future, the combination of pancreatic differentiation sys- reported to proliferate in the embryo and early childhood, adult β cell
tem, precise genome editing, and bioinformatics will help us con- replication is barely detectable [38]. The dysfunction of pancreatic β
struct more sophisticated diabetes-related modeling and investigate cells can cause type 2 diabetes, and screening chemical compounds
the underlying molecular mechanisms in more details. to protect and improve pancreatic β-cell function is in high demand.
For example, using a luciferase-based high-throughput chemical
screening strategy, Wang et al. [39] found that small molecules,
Applications of hPSC-derived Pancreatic β-like
harmine and INDY, functioned as a new class of human β cell
Cells in Drug Discovery mitogenic compounds. Mechanistically, they defined dual-specificity
The pharmaceutical industry in diabetes currently faces several chal- tyrosine-regulated kinase-1a (DYRK1A) as the possible target for
lenges, including the poor predictivity of efficacy and toxicity with harmine and INDY, and the nuclear factors of activated T cells
the preclinical biological models and the increasing costs of devel- family of transcription factors as the possible mediators of human
oping new drugs [37]. Promisingly, drug screening using hPSC- β-cell proliferation and function [39]. As a follow-up work, Wang
derived pancreatic β-like cells can address some of these challenges et al. [40] demonstrated that inhibition of DYRK1A, SMAD, and
(Fig. 3). Patient-specific pancreatic β-like cells carry many hallmarks Trithorax pathways could synergize to induce robust replication of
of genetic and epigenetic states and are suitable for both target- hPSC-derived pancreatic β-like cells and adult human pancreatic β
focused and phenotypic large-scale chemical screening. cells. Likewise, screening small molecules for pancreatic β-like cell
To date, several research groups have used isogenic hESC-derived differentiation, expansion, survival, and function will provide new
pancreatic β-like cells to functionally evaluate the GWAS-identified drug candidates for the treatment of diabetes.
6 Pancreatic β cells from hPSCs

It becomes more important to develop better differentiation They used hESC-derived RPE cell transplantation as an in vivo proof
protocols in a sufficiently robust and reproducible manner, as to of principle study and obtained very promising results. In the future,
meet the requirements of large-scale cell culture. In the next decades, similar approaches can also be applied for hPSC-derived pancreatic
additional work, including sensitive reporter systems, high-content β-like cell transplantation.

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
analysis, data mining, and machine learning, is needed to develop
better systems for drug discovery. Summary and Perspectives
Tremendous progresses have already been made to generate func-
Potential Applications of hPSC-derived tional pancreatic β-like cells from hPSCs. It is now possible to
Pancreatic β-like Cells in Cell-based Therapy create diabetes patient-specific iPSCs and their pancreatic derivatives.
Islet transplantation is an effective method for treating diabetes In the past decade, genome-editing approaches, such as CRISPR
but is severely limited by the scarceness of islet donors, the short tools, accelerated the detailed study of pancreatic differentiation and
time duration of insulin secretion after transplantation [41], and the function. Here, we reviewed recent advances on human pancreatic β-
adverse reactions caused by the use of immuno-suppressive drugs like cell differentiation from hPSCs and the potential applications of
[42]. Interestingly, it does not necessarily require the transplantation these hPSC-derived pancreatic β-like cells in disease modeling, drug
of mature pancreatic β cells. Human fetal pancreatic tissues can also discovery, and cell-based therapies.
develop functionally in mice [43] and human [44]. Obviously, one In the future, new technologies will emerge and be critically help-
major potential application of hPSC-derived pancreatic β-like cells ful to achieve these goals. More precise genome-editing tools can help
and tissues is for cell-based therapy (Fig. 4). to better study the underlying mechanisms of disease progression of
Immune rejection is a major obstacle for cell-based therapy, multiple forms of diabetes. Cost-effective high-throughput profiling
and better immune protection strategies are necessary to promote approaches can be used to better understand human pancreatic
more effective and longer lasting treatments. Stem cells with specific differentiation and development on multiple ‘omic’ levels, including
immunogenic modifications can allow them to escape from autoim- genomic, epigenomic, transcriptomic, proteomic, and metabolomic.
mune attacks in patients with type 1 diabetes [45]. For example, In addition, powerful high-throughput screening will provide new
Szot et al. [46] found that hESC-derived pancreatic endoderm cells drug candidates for effectively treating diabetes. By overcoming the
could survive in non-diabetic mice for more than 90 days using issues of immune rejection, safety, cost, and long-term functions, cell-
CTLA4Ig to inhibit T-cell activation and anti-CD154 antibody to based therapy will become a reality.
block the T-cell costimulatory pathway. Human leukocyte antigens Therefore, more support and investments for the young field of
(HLAs) on the cell surface are the main cause of CD4+ T-cell- stem cell biology should be provided by governments, academics,
mediated allograft rejection. In order to find a long-term solu- and industries. Very optimistically, these efforts will eventually lead
tion to the immune responses, many researchers tried to generate to significant advances in the field of regenerative medicine, provide
hypoimmunogenic hPSCs by genome editing. Lu et al. [47] disrupted innovative therapeutics, and benefit millions of people worldwide.
beta 2-microglobulin (β2m) and generated HLA class I-deficient
hESCs, which showed low immunogenicity. Chen et al. [48] found
that CIITA−/− hESCs could differentiate into cells with no HLA II Acknowledgement
expression. These cells can escape the attack of receptors’ CD4+ T We would like to thank all members of Zhu laboratory for helpful
cells. In addition, researchers from CRISPR Therapeutics generated discussions. Due to the space limitation, we apologize to those whose
genome-edited CyT49 cells that lack β2m and express PD-L1 to findings are relevant but not cited in this review.
further protect cells from T-cell attack [49].
Using cell impermeable macrocysts is another promising strategy
to solve the immune rejection of xenograft in diabetic patients [50]. Funding
Encapsulated with these devices, hESC-derived PPs could further
differentiate into pancreatic β-like cells in mice [51]. Another method This work was supported by the grants from National Natural
of islet encapsulation is to prepare the isolated islets into alginate Science Foundation of China (No. 31970818), the National Key
microspheres. The hESC-derived pancreatic β-like cells coated with Research and Development Program of China (No. 2016YFC1
triazolethiomorpholine dioxide alginate could regulate blood glucose 305300), and the Outstanding Youth Fund of Zhejiang Province
level and protect against immunosuppression for more than 170 days (No. R17C120002).
in mice with normal immunity [52]. Investigators at ViaCyte used the
CyT49 cells to generate PPs [53], and then these PPs could mature
into glucose-responsive and insulin-secreting pancreatic β-like cells References
in macroencapsulation devices in vivo [51]. Excitingly, PEC-Encap 1. Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson
clinical trial data for T1DM Phase I/II showed that the product was RP, Secchi A, et al. International trial of the Edmonton protocol for islet
safe and well tolerated, and the insulin-secreting cells could maintain transplantation. N Engl J Med 2006, 355: 1318–1330.
in vivo for more than 2 years [54]. Moreover, a potential advantage 2. Bruin JE, Rezania A, Kieffer TJ. Replacing and safeguarding pancreatic β
cells for diabetes. Sci Transl Med 2015, 7: 316–323.
of this approach is that the device can be more easily removed from
3. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S,
patients. Undoubtedly, these studies will set up the foundations for
Young H, et al. Pancreatic endoderm derived from human embryonic
further efforts to overcome immune rejection issues. stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat
For cell-based therapy, safety is critically required, and several Biotechnol 2008, 26: 443–452.
innovative and promising strategies are being tested in animal mod- 4. Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman
els. For example, Liang et al. [55] linked cell-division gene CDK1 M. Insulin production by human embryonic stem cells. Diabetes 2001,
to a suicide gene HSV-TK to define and improve cell therapy safety. 50: 1691–1697.
Pancreatic β cells from hPSCs 7

5. D’Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE. 26. Wei Z, Yoshihara E, He N, Hah N, Fan W, Pinto AFM, Huddy T, et al.
Efficient differentiation of human embryonic stem cells to definitive Vitamin D switches BAF complexes to protect beta cells. Cell 2018, 173:
endoderm. Nat Biotechnol 2005, 23: 1534–1541. 1135–1149.
6. D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart 27. Li QV, Dixon G, Verma N, Rosen BP, Gordillo M, Luo R, Xu C,

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
NG, Moorman MA, et al. Production of pancreatic hormone-expressing et al. Genome-scale screens identify JNK-JUN signaling as a barrier for
endocrine cells from human embryonic stem cells. Nat Biotechnol 2006, pluripotency exit and endoderm differentiation. Nat Genet 2019, 51:
24: 1392–1401. 999–1010.
7. Ma X, Zhu S. Chemical strategies for pancreatic beta cell differentiation, 28. Teo AK, Wagers AJ, Kulkarni RN. New opportunities: harnessing induced
reprogramming, and regeneration. Acta Biochim Biophys Sin 2017, 49: pluripotency for discovery in diabetes and metabolism. Cell Metab 2013,
289–301. 18: 775–791.
8. Chen S, Borowiak M, Fox JL, Maehr R, Osafune K, Davidow L, Lam K, 29. Flanagan SE, De Franco E, Lango Allen H, Zerah M, Abdul-Rasoul MM,
et al. A small molecule that directs differentiation of human ESCs into the Edge JA, Stewart H, et al. Analysis of transcription factors key for mouse
pancreatic lineage. Nat Chem Biol 2009, 5: 258–265. pancreatic development establishes NKX2-2 and MNX1 mutations as
9. Rezania A, Bruin JE, Riedel MJ, Mojibian M, Asadi A, Xu J, Gauvin causes of neonatal diabetes in man. Cell Metab 2014, 19: 146–154.
R, et al. Maturation of human embryonic stem cell-derived pancreatic 30. Zhu Z, Li QV, Lee K, Rosen BP, Gonzalez F, Soh CL, Huangfu D. Genome
progenitors into functional islets capable of treating pre-existing diabetes editing of lineage determinants in human pluripotent stem cells reveals
in mice. Diabetes 2012, 61: 2016–2029. mechanisms of pancreatic development and diabetes. Cell Stem Cell 2016,
10. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O’Dwyer 18: 755–768.
S, et al. Reversal of diabetes with insulin-producing cells derived in vitro 31. Zeng H, Guo M, Zhou T, Tan L, Chong CN, Zhang T, Dong X, et al. An
from human pluripotent stem cells. Nat Biotechnol 2014, 32: 1121–1133. isogenic human ESC platform for functional evaluation of genome-wide-
11. Pagliuca FW, Millman JR, Gurtler M, Segel M, Van Dervort A, Ryu JH, association-study-identified diabetes genes and drug discovery. Cell Stem
Peterson QP, et al. Generation of functional human pancreatic beta cells Cell 2016, 19: 326–340.
in vitro. Cell 2014, 159: 428–439. 32. Shi ZD, Lee K, Yang D, Amin S, Verma N, Li QV, Zhu Z, et al.
12. Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, Genome editing in hPSCs reveals GATA6 haploinsufficiency and a genetic
Guo T, et al. Controlled induction of human pancreatic progenitors pro- interaction with GATA4 in human pancreatic development. Cell Stem Cell
duces functional beta-like cells in vitro. EMBO J 2015, 34: 1759–1772. 2017, 20: 675–688 e676.
13. Wang W, Jin S, Ye K. Development of islet organoids from H9 human 33. Amin S, Cook B, Zhou T, Ghazizadeh Z, Lis R, Zhang T, Khalaj M,
embryonic stem cells in biomimetic 3D scaffolds. Stem Cells Dev 2017, et al. Discovery of a drug candidate for GLIS3-associated diabetes. Nat
26: 394–404. Commun 2018, 9: 2681.
14. Mamidi A, Prawiro C, Seymour PA, de Lichtenberg KH, Jackson A, 34. Zhou T, Kim TW, Chong CN, Tan L, Amin S, Sadat Badieyan Z,
Serup P, Semb H. Mechanosignalling via integrins directs fate decisions Mukherjee S, et al. A hPSC-based platform to discover gene-environment
of pancreatic progenitors. Nature 2018, 564: 114–118. interactions that impact human β-cell and dopamine neuron survival. Nat
15. Youngblood RL, Sampson JP, Lebioda KR, Shea LD. Microporous scaf- Commun 2018, 9: 4815.
folds support assembly and differentiation of pancreatic progenitors into 35. Lee K, Cho H, Rickert RW, Li QV, Pulecio J, Leslie CS, Huangfu D.
beta-cell clusters. Acta Biomater 2019, 96: 111–122. FOXA2 is required for enhancer priming during pancreatic differentia-
16. Velazco-Cruz L, Song J, Maxwell KG, Goedegebuure MM, tion. Cell Rep 2019, 28: 382–393 e387.
Augsornworawat P, Hogrebe NJ, Millman JR. Acquisition of dynamic 36. Cardenas-Diaz FL, Osorio-Quintero C, Diaz-Miranda MA, Kishore S,
function in human stem cell-derived β cells. Stem Cell Rep 2019, 12: Leavens K, Jobaliya C, Stanescu D, et al. Modeling monogenic diabetes
351–365. using human ESCs reveals developmental and metabolic deficiencies
17. Nair GG, Liu JS, Russ HA, Tran S, Saxton MS, Chen R, Juang C, et al. caused by mutations in HNF1A. Cell Stem Cell 2019, 25: 273–289 e275.
Recapitulating endocrine cell clustering in culture promotes maturation 37. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceu-
of human stem-cell-derived beta cells. Nat Cell Biol 2019, 21: 263–274. tical industry: new estimates of R&D costs. J Health Econ 2016, 47:
18. Toyoda T, Mae S, Tanaka H, Kondo Y, Funato M, Hosokawa Y, Sudo T, 20–33.
et al. Cell aggregation optimizes the differentiation of human ESCs and 38. Zhou Q, Melton DA. Pancreas regeneration. Nature 2018, 557: 351–358.
iPSCs into pancreatic bud-like progenitor cells. Stem Cell Res 2015, 14: 39. Wang P, Alvarez-Perez JC, Felsenfeld DP, Liu H, Sivendran S, Bender A,
185–197. Kumar A, et al. A high-throughput chemical screen reveals that harmine-
19. Yabe SG, Fukuda S, Takeda F, Nashiro K, Shimoda M, Okochi H. Effi- mediated inhibition of DYRK1A increases human pancreatic beta cell
cient generation of functional pancreatic beta-cells from human induced replication. Nat Med 2015, 21: 383–388.
pluripotent stem cells. J Diabetes 2017, 9: 168–179. 40. Wang P, Karakose E, Liu H, Swartz E, Ackeifi C, Zlatanic V, Wilson J,
20. Ghazizadeh Z, Kao DI, Amin S, Cook B, Rao S, Zhou T, Zhang T, et al. et al. Combined inhibition of DYRK1A, SMAD, and Trithorax pathways
ROCKII inhibition promotes the maturation of human pancreatic beta- synergizes to induce robust replication in adult human beta cells. Cell
like cells. Nat Commun 2017, 8: 298. Metab 2019, 29: 638–652 e635.
21. Ameri J, Borup R, Prawiro C, Ramond C, Schachter KA, Scharfmann R, 41. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL,
Semb H. Efficient generation of glucose-responsive beta cells from isolated Kneteman NM, et al. Islet transplantation in seven patients with type 1
GP2(+) human pancreatic progenitors. Cell Rep 2017, 19: 36–49. diabetes mellitus using a glucocorticoid-free immunosuppressive regimen.
22. Veres A, Faust AL, Bushnell HL, Engquist EN, Kenty JH, Harb G, Poh N Engl J Med 2000, 343: 230–238.
YC, et al. Charting cellular identity during human in vitro beta-cell 42. Lakey JR, Mirbolooki M, Shapiro AM. Current status of clinical
differentiation. Nature 2019, 569: 368–373. islet cell transplantation. In: Hornick P, Rose M eds. Transplantation
23. Alvarez-Dominguez JR, Donaghey J, Rasouli N, Kenty JHR, Helman A, Immunology: Methods and Protocols. Totowa, NJ: Humana Press, 2006,
Charlton J, Straubhaar JR, et al. Circadian entrainment triggers matura- 47–104
tion of human in vitro islets. Cell Stem Cell 2019, 5909: 30466–30467. 43. Tuch BE, Osgerby KJ, Turtle JR. Reversal of diabetes in hypergly-
24. Yu C, Liu Y, Ma T, Liu K, Xu S, Zhang Y, Liu H, et al. Small molecules caemic nude mice by human fetal pancreas. Transplant Proc 1989, 21:
enhance CRISPR genome editing in pluripotent stem cells. Cell Stem Cell 2665–2666.
2015, 16: 142–147. 44. Lafferty KJ, Hao L. Fetal pancreas transplantation for treatment of IDDM
25. Ma X, Chen X, Jin Y, Ge W, Wang W, Kong L, Ji J, et al. Small molecules patients. Diabetes Care 1993, 16: 383–386.
promote CRISPR-Cpf1-mediated genome editing in human pluripotent 45. Gjelberg HK, Hoem D, Verbeke CS, Eide J, Cooper JG, Molven A.
stem cells. Nat Commun 2018, 9: 1303. Hypoglycemia and decreased insulin requirement caused by malignant
8 Pancreatic β cells from hPSCs

insulinoma in a type 1 diabetic patient: when the hoof beats are from 51. Agulnick AD, Ambruzs DM, Moorman MA, Bhoumik A, Cesario RM,
a zebra, not a horse. Clin Case Rep 2017, 5: 761–768. Payne JK, Kelly JR, et al. Insulin-producing endocrine cells differentiated
46. Szot GL, Yadav M, Lang J, Kroon E, Kerr J, Kadoya K, Brandon EP, in vitro from human embryonic stem cells function in macroencapsulation
et al. Tolerance induction and reversal of diabetes in mice transplanted devices in vivo. Stem Cells Transl Med 2015, 4: 1214–1222.

Downloaded from https://academic.oup.com/abbs/advance-article-abstract/doi/10.1093/abbs/gmaa047/5842276 by Jamia Millia Islamia University user on 25 May 2020
with human embryonic stem cell-derived pancreatic endoderm. Cell Stem 52. Vegas AJ, Veiseh O, Gurtler M, Millman JR, Pagliuca FW, Bader AR,
Cell 2015, 16: 148–157. Doloff JC, et al. Long-term glycemic control using polymer-encapsulated
47. Lu P, Chen J, He L, Ren J, Chen H, Rao L, Zhuang Q, et al. Generating human stem cell-derived beta cells in immune-competent mice. Nat Med
hypoimmunogenic human embryonic stem cells by the disruption of beta 2016, 22: 306–311.
2-microglobulin. Stem Cell Rev Rep 2013, 9: 806–813. 53. Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, Bang AG,
48. Chen H, Li Y, Lin X, Cui D, Cui C, Li H, Xiao L. Functional disruption of Bhoumik A, et al. A scalable system for production of functional pancre-
human leukocyte antigen II in human embryonic stem cell. Biol Res 2015, atic progenitors from human embryonic stem cells. PLoS One 2012, 7:
48: 59. e37004.
49. Sluch V. CRISPR-editing of hESCs allows for production of immune 54. ViaCyte. Two-year data from ViaCyte’s STEP ONE clinical trial presented
evasive cells capable of differentiation to pancreatic progenitors for future at ADA 2018. Jun 23, 2018. https://viacyte.com/press-releases/two-year-
type 1 diabetes therapy. Sept 17, 2019. http://ir.crisprtx.com/static-files/ data-from-viacytes-step-one-clinical-trial-presented-at-ada-2018/.
af584c8b-5264-4bdd-a409-fec52e06d365. 55. Liang Q, Monetti C, Shutova MV, Neely EJ, Hacibekiroglu S, Yang H,
50. Desai T, Shea LD. Advances in islet encapsulation technologies. Nat Rev Kim C, et al. Linking a cell-division gene and a suicide gene to define and
Drug Discov 2017, 16: 338–350. improve cell therapy safety. Nature 2018, 563: 701–704.

You might also like