The Use of Animal Models to Study Stem Cell Therapies for Diabetes Mellitus

Page created by Jose Glover
 
CONTINUE READING
The Use of Animal Models to Study Stem Cell Therapies for Diabetes Mellitus

                                   Woo-Jin Song, Rohan Shah, and Mehboob A. Hussain

Abstract                                                                     solute (type 1) and a relative (type 2) reduction in functional
                                                                             insulin-producing β cell mass in the islets of Langerhans in
The two main forms of human diabetes mellitus (DM) are char-                 the pancreas. Type 1 DM results from autoimmune assault of

                                                                                                                                                Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
acterized by an absolute (type 1) and a relative (type 2) reduc-             β cells, and type 2 from the failure of pancreatic β cells to
tion in functional insulin-producing β cell mass in the pancreas.            sufficiently compensate for insulin resistance. Studies in
Type 1 DM results from autoimmune assault of β cells, and                    Europe indicate an increasing incidence of type 1 DM in
type 2 from the failure of pancreatic β cells to sufficiently com-           children in the past 15 years, and projections for 2005–2020
pensate for insulin resistance. Studies indicate that the inci-              estimate a doubling of new cases in children younger than
dence of both types is increasing rapidly to levels that constitute          5 years and an increase of approximately 70% in children
a global epidemic. Researchers are experimentally developing                 younger than 15 (Patterson et al. 2009). The incidence of
several conceptual approaches for increasing pancreatic β cell               type 2 DM is also increasing rapidly and is now considered
mass and testing them for feasibility in treating the disease. The           a global epidemic; in the United States there is an estimated
main sources for derivation of insulin-producing cells are em-               25-30% lifetime risk of developing the disease.
bryonic and induced pluripotent stem cells, endogenous pro-                      Efforts to develop treatments for the disease rely on
genitor cells (both within and outside the pancreas), stimulation            chemically induced animal (primarily rodent) models in
of β cell proliferation, and genetic “reprogramming” of cells.               which endogenous β cells are destroyed, most often by phar-
Strategies to effectively address immune- and inflammation-                  macologic toxin administration. β cell destruction results in
mediated assault on existing and newly formed β cells need to                lack of insulin production and pronounced hyperglycemia,
be refined. This review provides a description of β cell ablation            which, if untreated, leads to the death of the animal. Insulin-
methods and a discussion of various types of studies of regen-               producing cells, derived from a variety of sources, are tested
erative approaches—β cell proliferation, islet cell transplanta-             in these diabetic animals to assess the extent of recovery
tion, transdifferentiation, and the use of embryonic and induced             from hyperglycemia.
pluripotent stem cells—to the treatment of diabetes mellitus.                    Although the derivation of insulin-producing cells from
Although there has been much progress in this area, further re-              precursor cells is apparently becoming a feasible approach,
search is needed to enhance understanding and improve thera-                 considerable challenges remain in the treatment of diabetes
peutic strategies for this widespread disease.                               mellitus. For example, it is unclear whether newly generated
                                                                             β cells may form tumors and/or be able to function as well as
Key Words: ablation; β cell; diabetes mellitus; progenitor;                  endogenous β cells in the islets of Langerhans. The studies
proliferation; reprogramming; stem cells; transdifferentiation               described below attempt to address these and other questions
                                                                             about possible DM treatment strategies.
Introduction
                                                                             β Cell Ablation Methods

D
         iabetes mellitus (DM1) is increasingly widespread in
         the United States and Europe. The two main forms
                                                                             In vivo functional testing of insulin-producing cells is per-
         of the disease in humans are characterized by an ab-
                                                                             formed in animals whose endogenous β cells have been ab-
                                                                             lated, either partially (for studies of regeneration from
                                                                             residual β cells) or completely (for studies of de novo gen-
Woo-Jin Song, MS, is a graduate student in the Graduate Program in
Biological Chemistry in the Department of Pediatrics; Rohan Shah is an
                                                                             eration of β cells).
undergraduate student in the Biomedical Engineering Department; and
Mehboob A. Hussain, MD, is an associate professor in the Departments of
Pediatrics, Medicine, and Biological Chemistry, all at the Johns Hopkins     Chemical Ablation
University in Baltimore, Maryland.
    Address correspondence and reprint requests to Dr. Mehboob A. Hussain,   The two most popular chemicals for pharmacologic β cell ab-
Metabolism Division, Departments of Pediatrics, Medicine, and Biological     lation are streptozotocin (STZ1) and alloxan (Elsner et al.
Chemistry, Johns Hopkins University, 600 N. Wolfe Street, CMSC 10-113,
Baltimore, MD 21287 or email mhussai4@gw.johnshopkins.edu.
                                                                             2006). STZ, a nitrosourea analogue, decomposes into glucose
    1Abbreviations used in this article: DM, diabetes mellitus; GLUT2,       and a methylnitrosourea moiety that serves as an alkylating
glucose transporter 2; PPX, partial pancreatectomy; STZ, streptozotocin      agent, which fragments DNA, modifies macromolecules,

74                                                                                                                               ILAR Journal
and results in the destruction of β cells. STZ also damages        Differentiation of Pancreatic Duct Epithelium and Partial
mitochondrial function in part by alkylating and destroying        Duct Ligation Studies provides details.
mitochondrial DNA, a result that is evident in reduced
glucose-stimulated insulin secretion and that also contributes
to β cell demise (Lenzen 2008). STZ administration does not        Genetic Models of β Cell Ablation
prevent pancreas regeneration after partial pancreatectomy in
                                                                   Concerns about the toxicity of STZ and alloxan to GLUT2-
rats (Finegood et al. 1999).
                                                                   expressing cells that may be possible progenitors for β cell
    Alloxan generates reactive oxygen species (ROS) in a cy-
                                                                   regeneration have led to the creation of genetic models that
clic oxidation-reduction (redox) reaction in the presence of
                                                                   allow targeted ablation of insulin-expressing cells only. One
intracellular thiols (e.g., glutathione). The resulting molecule
                                                                   model is the PANIC-ATTAC2 transgenic mouse model, in
is dialuric acid, which via autooxidation generates superox-
                                                                   which β cell–specific and dose-dependent pharmacological
ide radicals, hydrogen peroxide, and hydroxyl radicals. In ad-
                                                                   activation of the cellular apoptosis cascade results in vari-
dition, alloxan inhibits glucokinase, which functions as the

                                                                                                                                                Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
                                                                   able degrees of β cell ablation (Wang et al. 2008). These
glucose sensor for the β cell, thereby suppressing glucose-
                                                                   mice reveal in their islets a population of cells that express
stimulated insulin secretion (Lenzen 2008). Both STZ and
                                                                   GLUT2 but not insulin, thus supporting the hypothesis that
alloxan are toxic glucose analogues that accumulate in pan-
                                                                   GLUT2-positive, insulin-negative cells may represent a
creatic β cells after entering them via the glucose transporter
                                                                   source for the formation of new functional β cells (Guz et al.
2 (GLUT21) (Elsner et al. 2002), which the cells express at
                                                                   2001; Wang et al. 2008).
high levels. However, other cell types also express GLUT2
                                                                       A second model of inducible β cell ablation is the β cell–
and the toxic effects of STZ on these cells have been a con-
                                                                   specific conditional expression of diphtheria toxin A (DTA)
cern in regenerative studies. Other experimental paradigms
                                                                   in genetically engineered mice (Nir et al. 2007). Mice do not
indicate the existence of an insulin-negative, GLUT2-positive
                                                                   naturally express the diphtheria toxin receptor, thus protect-
cell population in the islets of Langerhans, which may pro-
                                                                   ing all cells from DTA except those that express inducible
vide the cellular substrate for β cell regeneration (or matura-
                                                                   DTA. In this model, replication (i.e., mitosis of existing, re-
tion) (Guz et al. 2001; Wang et al. 2008). These observations
                                                                   sidual β cells) is the predominant if not sole mechanism of
are not mutually inconsistent. Rather, the different models
                                                                   regeneration after partial β cell destruction (Nir et al. 2007).
may indicate the possibility of β cell regeneration from differ-
ent cell compartments of the pancreas, of which some cell
populations may actually be intermediary derivations from          β Cell Proliferation Studies
other cell populations. For example, cells that give rise to β
cells in the partial pancreatectomy model (described in the        Proliferation of β cells occurs during developmental forma-
next section) may actually start off as GLUT2-negative and         tion of the pancreas and, in rodents, during the early postna-
thus be relatively protected from STZ; during the downstream       tal period, concurrent with a wave of apoptosis and
process toward differentiation to β cells, the differentiating     proliferation (Bonner-Weir 2000; Georgia and Bhushan
cells express GLUT2 and thus become vulnerable to STZ.             2004). In addition, functional adaptation of pancreatic β cells
                                                                   to increasing insulin requirements, again in rodents, likely
Surgical Ablation of Pancreatic                                    occurs primarily as a result of proliferation, the primary
Tissue (Partial Pancreatectomy)                                    mechanism of β cell mass adaptation to metabolic demands.
                                                                   Typical circumstances of increased insulin demand are diet-
Partial pancreatectomy (PPX1; removal of 60-90% of the             induced insulin resistance and pregnancy, when the failure
pancreatic corpus and tail) in rodents is followed by a robust     of β cell mass adaptation to metabolic demands results, re-
regeneration of pancreatic tissue, including the endocrine         spectively, in impaired glucose tolerance or frank type 2 DM
compartment, that is complete approximately 2 weeks after          and gestational DM.
the procedure (Bonner-Weir et al. 1993). Studies indicate              Genomewide association studies have linked type 2 DM
that most if not all of the regenerating β cells derive from       to single-nucleotide polymorphisms in genes, which are
proliferation of existing β cells (Dor et al. 2004). But careful   highly expressed in β cells. The products of a number of
histological assessment has also revealed a proliferation of       these identified genes are proteins, which regulate the cell
epithelial cells in small ductules in the pancreas, and these      cycle (Saxena et al. 2007; Scott et al. 2007). Observations of
cells give rise to newly formed islets, pancreatic lobules, and    β cell proliferation associated with increased metabolic de-
acinar tissue (Bonner-Weir et al. 1993).                           mands in rodents, combined with the genomewide associa-
    Partial pancreatic duct ligation in rodents involves place-    tion studies of type 2 DM, have led to the hypothesis that
ment of a stricture (usually across the middle or proximal         type 2 DM in humans results in part from disturbed β cell
third of the pancreas) to obstruct the pancreatic duct, leaving    mass adaptation (i.e., proliferation).
the vascular pancreatic perfusion intact. The ligation causes
the rapid atrophy of tissue distal to the ligation and the
proliferation of duct epithelium, which gives rise to newly        2PANIC-ATTAC stands for pancreatic islet β cell apoptosis through targeted

formed islets (neogenesis) and acinar tissue; the section on       activation of caspase 8.

Volume 51, Number 1       2010                                                                                                           75
β Cell Proliferation and Diet-Induced Obesity                       β cells have an equal capacity to proliferate (Brennand et al.
                                                                    2007; Teta et al. 2007).
A robust model of increased peripheral and hepatic insulin
resistance entails a high-fat diet (60% of calories in the
form of lipids) for mice, which develop obesity and insulin         Decline in β Cell Proliferation Capacity
resistance accompanied by β cell proliferation. Depending           with Aging
on the strain of mice, either β cell proliferation is robust and
the animals do not develop diabetes, or proliferation is            β cell proliferation is robust in young rodents (in mice up to
insufficient to meet metabolic demands and the animals              approximately 2 months of age), but aging is accompanied
develop glucose intolerance and a type 2 DM phenotype.              by diminished capacity of β cells to proliferate in response
Studies have exploited differences between mouse strains            to various physiological, pharmacological, or experimental
to assess gene expression profiles in various tissues (Keller       stimuli (Rankin and Kushner 2009; Tschen et al. 2009). Ge-
et al. 2008), with prominent differences in expression of           netic studies of the restriction of β cell proliferation indicate

                                                                                                                                         Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
cell cycle regulators in the islets of a mouse strain prone to      a prominent role for the cell cycle negative regulatory pro-
developing diabetes with diet-induced obesity versus one            teins encoded by the Ink4/Arf locus. Ablation of p16INK4a
that is not.                                                        results in preserved β cell regeneration after STZ-induced
                                                                    partial β cell ablation in aging mice (Krishnamurthy et al.
                                                                    2006). The Ink4/Arf locus is subject to epigenetic regulation
β Cell Mass Adaptation During Pregnancy                             by Polycomb proteins with age-related increased p16 ex-
                                                                    pression and repression of β cell proliferation (Chen et al.
β cell mass adaptation during pregnancy occurs via β cell           2009; Dhawan et al. 2009).
proliferation. The current understanding is that during gesta-          Given these observations, it is reasonable to expect that
tion placental lactogen and leptin, which are produced in           young rodents may not reflect the circumstances of adult hu-
high quantities by the placenta, stimulate β cell proliferation.    mans, in which β cell proliferation, although documented, is
Intracellular signaling for placental lactogen occurs via pro-      rare and not as robust as in the early postnatal period (Butler
lactin receptors, which, together with leptin receptors, are        et al. 2007b; Meier et al. 2008). In addition, studies combin-
expressed on β cells and stimulate the intracellular JAK2/          ing the immunohistochemistry of human cadaver pancreas
STAT signaling pathway. In the case of the prolactin recep-         tissue and the mathematical modeling of β cell mass suggest
tor, JAK2/STAT5 signaling results in reduced expression of          that in humans β cells form throughout adult life not from
the tumor suppressor menin (Festa et al. 1999; Freemark et al.      the duplication of existing β cells but rather from as yet un-
2002; Huang et al. 2009; Karnik et al. 2005, 2007; Vasavada         identified non-β cells (e.g., ductal epithelial cells, acinar
et al. 2000; Yamashita et al. 2001).                                cells, centroacinar cells, nonpancreas resident cells) (Saisho
     Circulating serotonin levels increase and enzymes for          et al. 2009).
serotonin synthesis are upregulated in mouse islets during
gestation (Kim et al. 2009; Schraenen et al. 2009). Impor-
tantly, inhibition of serotonin production by pharmacologic         Transdifferentiation Studies
agents in pregnant mice as well as in β cell lines in vitro         and Reprogramming
markedly reduces β cell proliferation (Kim et al. 2009). Thus
serotonin synthesis by β cells appears to be an important step      To address circumstances when no β cells are present (e.g., in
in their adaptation during pregnancy. Further research is nec-      long-standing type 1 diabetes) or existing β cells do not pro-
essary to determine how β cell serotonin is linked to the           liferate (see above), researchers have tested the possibility of
stimulation of cell cycle progression.                              replacing β cells through the “transdifferentiation” of non-β
                                                                    cells from a differentiated phenotype to a β cell phenotype
                                                                    using a variety of cell sources. Among these are cells from
Methods of Inducing Cell Proliferation                              the pancreas (e.g., pancreatic duct epithelial cells or acinar
                                                                    cells) or extrapancreatic tissue (e.g., liver, bone marrow). In
In addition to “physiological” circumstances of growth, diet-
                                                                    the past 20 years, the discovery of β cell growth factors and
induced insulin resistance, and pregnancy, β cell prolifera-
                                                                    transcription factors, which regulate pancreas and β cell de-
tion can be stimulated in rodents and observed after partial
                                                                    velopment and differentiation, has provided important tools
ablation of β cell mass (i.e., through the administration of
                                                                    for gene delivery and novel genetically engineered animal
alloxan, STZ, inducible apoptosis, or DTA expression) or
                                                                    models that permit targeted investigation of the concept of
PPX (Dor et al. 2004). In addition, the incretin hormone
                                                                    transdifferentiation of non-β cells to functional β cells.
glucagon-like peptide 1 (GLP-1) and its long-acting peptide
analogue exendin-4 stimulate β cell proliferation in rodents
via the GLP-1 receptor, which is expressed at high levels on        Differentiation of Pancreatic Duct Epithelium
β cells (Song et al. 2008; Stoffers et al. 2000; Xu et al. 1999).
Studies that involve both the labeling of proliferating cells       Pancreatic duct epithelium has long been considered to har-
and genetic lineage tracing indicate that in mouse models all       bor cells with the capacity to differentiate into pancreatic

76                                                                                                                        ILAR Journal
endocrine cells under experimental circumstances. Rodent                 In vivo expression of a combination of developmentally
and human pancreatic duct epithelium can be cultured in              important transcription factors (pdx-1, mafA, ngn3) in pan-
vitro and differentiated into islet-like structures that, after      creatic acinar cells by adenovirus transduction results in a
transplantation, can reverse diabetes in a mouse lacking β           reprogramming of acinar cells into insulin-secreting β cells
cells (Bonner-Weir et al. 2000, 2004, 2008; Hao et al. 2006;         and rapid, sustained recovery from STZ-induced diabetes in
Inada et al. 2008). Studies combining genetic lineage tracing        mice (Zhou et al. 2008). The reprogrammed cells exhibit ul-
of pancreatic duct epithelial cells with partial duct ligation       trastructural features of pancreatic β cells and also attract
reveal the capacity for these cells to differentiate into pan-       newly formed capillary blood vessels by expressing vascular
creatic acinar and endocrine cells (Bonner-Weir et al. 2008;         endothelial growth factor (VEGF), which β cells have also
Inada et al. 2008).                                                  been shown to express (Zhou et al. 2008).
    Additional studies indicate evidence of a subpopulation
of pancreatic duct epithelial cells that, after partial duct liga-
tion, express the developmental transcription factor neuro-          β Cell Derivation from Liver Cells

                                                                                                                                        Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
genin 3 (ngn3) and contribute to newly formed endocrine
                                                                     The conversion of hepatocytes into pancreatic endocrine
cells. Neurogenin 3 is critical for pancreatic endocrine devel-
                                                                     cells with reversal of STZ-induced diabetes in rodents has
opment, as shown by the complete lack of pancreatic endo-
                                                                     been reported by several groups that have used similar strate-
crine cells in ngn3-null mice (Gradwohl et al. 2000) and the
                                                                     gies of gene delivery and have reprogrammed hepatocytes to
lack of neogenesis of such cells after duct ligation (Xu et al.
                                                                     β cell–like cells. The common feature in these studies is the
2008). A recent study demonstrated that pancreatic duct epi-
                                                                     delivery of either the transcription factor pdx-1, alone or in
thelial cells differentiate into glucagon-expressing β cells
                                                                     combination (Ber et al. 2003; Ferber et al. 2000) with beta-
(Collombat et al. 2009): when the paired homeodomain tran-
                                                                     cellulin (Kojima et al. 2003), or a modified pdx-1 cDNA that
scription factor pax4 is misexpressed in the newly formed β
                                                                     results in enhanced transcriptional activity of pdx-1 (Li et al.
cells, they differentiate into a β cell phenotype such that newly
                                                                     2005a; Thowfeequ et al. 2009).
formed β cells functionally reverse chemically induced DM.
                                                                         Researchers have recently shown that ngn3 transduction
    In contrast, genetic lineage tracing studies in mice indi-
                                                                     into liver cells results not only in transient expression of in-
cate that pancreatic endocrine regeneration after PPX occurs
                                                                     sulin in hepatocytes but also, more importantly, in the emer-
primarily via proliferation of existing β cells (Dor et al.
                                                                     gence of periportal clusters of islet-like cells that express
2004) and does not require reexpression of ngn3 (Lee et al.
                                                                     phenotypic markers of islet cells, allowing for long-term re-
2006). These studies suggest differences between the mo-
                                                                     versal of STZ-induced diabetes in mice (Yechoor et al.
lecular mechanisms underlying pancreatic endocrine cell
                                                                     2009). The authors propose that progenitor cells in the peri-
(re)generation after PPX versus partial duct ligation, with
                                                                     portal structures in the liver can, via ngn3 transduction, be
pancreatic duct epithelium cells possessing the capacity to
                                                                     “transdetermined” to differentiation into pancreatic islet-like
reexpress ngn3 and possibly to recapitulate ontogenic steps
                                                                     structures (Yechoor et al. 2009).
of pancreatic endocrine differentiation. When comparing re-
covery of β cell mass after specific experimental means of
reduction (β cell ablation, PPX, and pancreatic duct liga-           Cells from Bone Marrow
tion), it appears that the different types of injury result in
recruitment of different cell types in the pancreas to regener-      A small number of studies have reported the ability of bone
ate pancreatic endocrine mass.                                       marrow–derived cells to differentiate into insulin-producing
                                                                     cells after transplantation in animals (Hess et al. 2003; Ianus
                                                                     et al. 2003; Wang et al. 2006). STZ diabetic mice that re-
Pancreatic Acinar Cells and Reprogramming                            ceived a transplant of a bone marrow–derived c-kit+ cell
                                                                     population recovered from their diabetes. While a small frac-
The exocrine pancreatic acinar cells (Minami et al. 2005;            tion (1-3%) of donor-derived cells are insulin-positive in the
Okuno et al. 2007) have been differentiated in vitro to pancre-      pancreas, the main observation is that the transplantation
atic endocrine β cells through the overexpression of transcrip-      procedure itself appears to stimulate host β cell proliferation,
tion factors, which are important for β cell differentiation and     which is likely the main reason for the rapid recovery of host
phenotype maintenance (Li et al. 2005b; Wang et al. 2009).           β cell mass and reversal of the diabetes phenotype (Hess et al.
    In vivo delivery of pdx-1 and betacellulin (a peptidergic        2003). Other studies have failed to show evidence of bone
β cell differentiation factor) into the acinar tissue of STZ dia-    marrow cell differentiation into pancreatic endocrine cells
betic rats, through transabdominal ultrasound-guided dis-            (Akashi et al. 2008; Choi et al. 2003; Lechner et al. 2004).
ruption of the microvesicles (and from there into the                    In humans, maternal microchimerism (as a result of
circulation), permanently reverses the diabetes phenotype.           maternal-to-fetal transfer of circulating blood cells during
Histological analysis indicates that the pancreatic acinar           parturition) leads to female cells in male offspring. A recent
cells are converted into cells with certain characteristics of β     study indicates that β cells with a female chromosomal com-
cells while acinar markers also remain detectable in these cells     plement are present in male islets, suggesting the possibility
(Chen et al. 2007).                                                  of bloodborne cells, which under certain circumstances have

Volume 51, Number 1       2010                                                                                                    77
the capacity to engraft islets and express a pancreatic endo-       gies for improving iPS cell generation include replacing
crine phenotype (Nelson et al. 2007). In contrast, autopsy          some if not all of the reprogramming transcription factors
studies of hematopoietic stem cell transplant recipients reveal     with small molecular compounds and increasing the effi-
no evidence of pancreatic endocrine engraftment by donor            ciency of iPS cell generation in an initial pool of cells
cells (Butler et al. 2007a). Nevertheless, clinical trials on the   (Huangfu et al. 2008a,b). As with ESCs, it is important to
basis of these observations have reported a reversal of clinical    address the possibility of teratoma formation, which is a
type 1 diabetes in humans after autologous treatment with           property of iPS cells, before considering large-scale in vivo
bone marrow cell transplantation (Voltarelli et al. 2008).          clinical applications.
    These initial studies appear encouraging, but it remains            iPS cells have been differentiated along lineages of a va-
unclear whether bone marrow harbors a cell type that may            riety of tissue-specific phenotypes, include neuronal and car-
have the capacity to differentiate into a pancreatic endocrine      diac phenotypes (Carey et al. 2009; Hanna et al. 2008;
cell phenotype. The lack of a mechanistic cellular and mo-          Stadtfeld et al. 2008a). This variety opens the possibility of
lecular understanding of these observations certainly indi-         generating disease-specific iPS cells that can be differenti-

                                                                                                                                        Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
cates the need for further elucidation.                             ated for detailed in vitro studies (Park et al. 2008). A recent
                                                                    report has described in vitro differentiation of insulin-
                                                                    producing cells from human iPS cells (Zhang et al. 2009); so
Studies with Embryonic Stem Cells                                   far, there are no reports of transplantation studies of iPS-
                                                                    derived glucose-sensing and insulin-secreting cells.
Molecular mechanisms governing development of the pan-
creas and endocrine pancreas have provided insight and tools
for directed differentiation of human and mouse embryonic           Islet and Islet-like Cell
stem cells (ESCs) to glucose-responsive insulin-producing           Transplantation Studies
cells with close phenotypical resemblance to pancreatic β
                                                                    In vitro–generated β cells, β cell surrogates, or islet-like
cells. Protocols for ESC-to-β cell differentiation have been
                                                                    structures can be tested for their response to a glucose stimu-
developed by attempting to stimulate the signaling cascades
                                                                    lus by in vitro exposure to various secretagogue stimuli, of
that govern embryonic β cell development and differentiation,
                                                                    which glucose is the primary physiological stimulant. Static
resulting in recapitulation of the developmental transcription
                                                                    incubation of β cell surrogates allows an assessment of dose-
signature of pancreas endocrine and β cell differentiation.
                                                                    dependent secretion of insulin and C-peptide, both of which
A critical initial step is the differentiation into endoderm by
                                                                    accumulate in the culture supernatant over time and can be
exposure of ESCs to the Activin A analogue Nodal. A step-
                                                                    determined with a standard enzyme-linked immunosorbent
wise exposure to a variety of differentiation factors results
                                                                    assay (ELISA). Dynamic response to secretagogues can be
in differentiation of cells toward a β cell phenotype
                                                                    assessed by perifusion of islet-like structures and β cell sur-
(D’Amour et al. 2006; Kroon et al. 2008); recent reviews
                                                                    rogates (perifusion involves the incubation of the test tissue
summarize protocols for differentiating ESCs into β cell
                                                                    in a perifusion chamber, where the tissue is exposed to con-
phenotypes (Oliver-Krasinski and Stoffers 2008; Spence
                                                                    trolled dynamic changes of secretagogues; dynamic changes
and Wells 2007).
                                                                    in insulin and C-peptide can be measured in the effluent
    Glucose-responsive insulin-secreting cells derived from
                                                                    from the chamber) (Hussain et al. 2006).
ESCs have been tested in vivo after transplantation in dia-
                                                                         Transplantation of insulin-producing cells, islets, and is-
betic mice. Although glucose metabolism can be satisfactorily
                                                                    let-like structures permits in vivo assessment of the functional
controlled, the efficiency of successful ESC differentiation
                                                                    performance of newly derived β cells. Before transplantation,
to β cells is low (approximately 10% of ESCs differentiate
                                                                    syngeneic (to avoid immune rejection) or immunocompro-
into glucose-responsive cells) and potential teratoma forma-
                                                                    mised recipient animals are rendered diabetic by β cell abla-
tion from ESC-derived tissue poses safety concerns for clini-
                                                                    tion using STZ. Sites for transplantation include the peritoneal
cal application.
                                                                    cavity, the portal vein (which introduces islets into the low-
                                                                    pressure hepatic perfusion system), and a hollow space cre-
Studies with Induced Pluripotent Stem Cells                         ated between the kidney capsule and the kidney parenchyma.
                                                                    The space under the kidney capsule is most commonly used
Induced pluripotent stem (iPS) cells were originally derived        and provides a confined, accessible area for transplantation
from cultured mouse fibroblasts by stably or transiently over-      while also permitting adequate blood and nutrient supply to
expressing three transcription factors (Oct4, Sox2, Klf4)           the newly transplanted tissue. Depending on glucose sensing,
highly enriched in pluripotent ESCs (Stadtfeld et al. 2008b;        insulin synthesis, and the secretion properties and capacity of
Takahashi and Yamanaka 2006; Yu et al. 2009). Other studies         the transplanted tissue, glucose levels begin to decrease in the
with the same transcription factors enabled the generation of       formerly diabetic transplant recipient. One advantage of
iPS cells from human fibroblasts (Takahashi et al. 2007) and        transplantation under the kidney capsule is that after in vivo
the reprogramming of terminally differentiated somatic              testing, the transplanted insulin-producing tissue can be re-
cells, including human β cells, to a pluripotent state (Carey       moved to assess whether glucose levels return to diabetic lev-
et al. 2009; Hanna et al. 2008; Stadtfeld et al. 2008a). Strate-    els in the surviving transplant host animal.

78                                                                                                                       ILAR Journal
In an attempt to move toward allo- and xenotransplanta-                      extended liver to pancreas transdifferentiation. J Biol Chem 278:31950-
tion of glucose-responsive insulin-producing tissue across                       31957.
                                                                              Bonner-Weir S. 2000. Life and death of the pancreatic beta cells. Trends
species and across immunologically restricted barriers, encap-                   Endocrinol Metab 11:375-378.
sulation techniques now enable nutrients to enter and reach                   Bonner-Weir S, Baxter LA, Schuppin GT, Smith FE. 1993. A second path-
the encapsulated tissue and insulin and C-peptide to exit the                    way for regeneration of adult exocrine and endocrine pancreas. A
capsule, while the capsule barrier prevents cellular interaction                 possible recapitulation of embryonic development. Diabetes 42:
of the host immune system. Of the large variety of substances                    1715-1720.
                                                                              Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song KH, Sharma A,
tested for encapsulation purposes, alginate is the enduring                      O’Neil JJ. 2000. In vitro cultivation of human islets from expanded duc-
main component that allows effective diffusion of glucose to                     tal tissue. Proc Natl Acad Sci U S A 97:7999-8004.
reach the encapsulated tissue and insulin to exit the capsule                 Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, Aye T, Sharma A.
and enter the host circulation (Barnett et al. 2006, 2007).                      2004. The pancreatic ductal epithelium serves as a potential pool of pro-
                                                                                 genitor cells. Pediatr Diabetes 5 Suppl 2:16-22.
                                                                              Bonner-Weir S, Inada A, Yatoh S, Li WC, Aye T, Toschi E, Sharma A. 2008.
Concluding Remarks and Outlook

                                                                                                                                                             Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
                                                                                 Transdifferentiation of pancreatic ductal cells to endocrine beta-cells.
                                                                                 Biochem Soc Trans 36:353-356.
There has been tremendous progress in the past decade to-                     Brennand K, Huangfu D, Melton D. 2007. All beta cells contribute equally
                                                                                 to islet growth and maintenance. PLoS Biol 5:e163.
ward the generation of glucose-sensing and insulin-secreting                  Butler AE, Huang A, Rao PN, Bhushan A, Hogan WJ, Rizza RA, Butler PC.
cells from a variety of cell sources. Animal models of diabe-                    2007a. Hematopoietic stem cells derived from adult donors are not a
tes mellitus will become increasingly important to studies                       source of pancreatic beta-cells in adult nondiabetic humans. Diabetes
of the multitude of progenitor cell–derived insulin-producing                    56:1810-1816.
cells. With respect to type 1 DM, recurrence of autoimmu-                     Butler PC, Meier JJ, Butler AE, Bhushan A. 2007b. The replication of beta
                                                                                 cells in normal physiology, in disease and for therapy. Nat Clin Pract
nity against new β cells will have to be addressed in conjunc-                   Endocrinol Metab 3:758-768.
tion with replacing the (destroyed) β cells. Humanized mouse                  Carey BW, Markoulaki S, Hanna J, Saha K, Gao Q, Mitalipova M, Jaenisch
models that mimic the immune system of human autoim-                             R. 2009. Reprogramming of murine and human somatic cells using a
mune disease would be an ideal model for future studies.                         single polycistronic vector. Proc Natl Acad Sci U S A 106:157-162.
    In type 2 DM, the fundamental underlying metabolic de-                    Chen H, Gu X, Su IH, Bottino R, Contreras JL, Tarakhovsky A, Kim SK.
                                                                                 2009. Polycomb protein ezh2 regulates pancreatic beta-cell ink4a/arf ex-
fects are not fully understood. Although a relative deficiency
                                                                                 pression and regeneration in diabetes mellitus. Genes Dev 23:975-985.
of β cells is characteristic of the disease, it remains unclear               Chen S, Ding J, Yu C, Yang B, Wood DR, Grayburn PA. 2007. Reversal of
whether increasing functional pancreatic β cell mass can be                      streptozotocin-induced diabetes in rats by gene therapy with betacellu-
an adequate and feasible treatment strategy. Further studies                     lin and pancreatic duodenal homeobox-1. Gene Ther 14:1102-1110.
in animal models are necessary to determine how increases                     Choi JB, Uchino H, Azuma K, Iwashita N, Tanaka Y, Mochizuki H, Migita
                                                                                 M, Shimada T, Kawamori R, Watada H. 2003. Little evidence of trans-
in β cell mass and insulin-secretion capacity might affect the
                                                                                 differentiation of bone marrow-derived cells into pancreatic beta cells.
metabolic milieu of preexisting type 2 DM with insulin re-                       Diabetologia 46:1366-1374.
sistance and dyslipidemia.                                                    Collombat P, Xu X, Ravassard P, Sosa-Pineda B, Dussaud S, Billestrup N,
                                                                                 Madsen OD, Serup P, Heimberg H, Mansouri A. 2009. The ectopic ex-
                                                                                 pression of pax4 in the mouse pancreas converts progenitor cells into
Acknowledgments                                                                  alpha and subsequently beta cells. Cell 138:449-462.
                                                                              D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG,
MAH is funded by the National Institutes of Health (grants                       Moorman MA, Kroon E, Carpenter MK, Baetge EE. 2006. Production
                                                                                 of pancreatic hormone-expressing endocrine cells from human embry-
RO1 DK 64646, RO1 DK 81472, RO1 DK 63349, RO1 DK
                                                                                 onic stem cells. Nat Biotechnol 24:1392-1401.
63349-05A1S1 [ARRA]) and the Baltimore Diabetes Re-                           Dhawan S, Tschen SI, Bhushan A. 2009. Bmi-1 regulates the ink4a/arf lo-
search and Training Center (P60 DK 079637).                                      cus to control pancreatic beta-cell proliferation. Genes Dev 23:906-
                                                                                 911.
                                                                              Dor Y, Brown J, Martinez OI, Melton DA. 2004. Adult pancreatic beta-cells
References                                                                       are formed by self-duplication rather than stem-cell differentiation. Na-
                                                                                 ture 429:41-46.
Akashi T, Shigematsu H, Hamamoto Y, Iwasaki H, Yatoh S, Bonner-Weir S,        Elsner M, Tiedge M, Guldbakke B, Munday R, Lenzen S. 2002. Importance
   Akashi K, Weir GC. 2008. Bone marrow or foetal liver cells fail to in-        of the glut2 glucose transporter for pancreatic beta cell toxicity of al-
   duce islet regeneration in diabetic Akita mice. Diabetes Metab Res Rev        loxan. Diabetologia 45:1542-1549.
   24:585-590.                                                                Elsner M, Gurgul-Convey E, Lenzen S. 2006. Relative importance of cel-
Barnett BP, Kraitchman DL, Lauzon C, Magee CA, Walczak P, Gilson WD,             lular uptake and reactive oxygen species for the toxicity of alloxan and
   Arepally A, Bulte JW. 2006. Radiopaque alginate microcapsules for x-          dialuric acid to insulin-producing cells. Free Radic Biol Med 41:825-
   ray visualization and immunoprotection of cellular therapeutics. Mol          834.
   Pharm 3:531-538.                                                           Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I,
Barnett BP, Arepally A, Karmarkar PV, Qian D, Gilson WD, Walczak P,              Seijffers R, Kopolovic J, Kaiser N, Karasik A. 2000. Pancreatic and
   Howland V, Lawler L, Lauzon C, Stuber M, Kraitchman DL, Bulte JW.             duodenal homeobox gene 1 induces expression of insulin genes in liver
   2007. Magnetic resonance-guided, real-time targeted delivery and im-          and ameliorates streptozotocin-induced hyperglycemia. Nat Med 6:568-
   aging of magnetocapsules immunoprotecting pancreatic islet cells. Nat         572.
   Med 13:986-991.                                                            Festa A, Shnawa N, Krugluger W, Hopmeier P, Schernthaner G, Haffner
Ber I, Shternhall K, Perl S, Ohanuna Z, Goldberg I, Barshack I, Benvenisti-      SM. 1999. Relative hypoleptinaemia in women with mild gestational
   Zarum L, Meivar-Levy I, Ferber S. 2003. Functional, persistent, and           diabetes mellitus. Diabet Med 16:656-662.

Volume 51, Number 1           2010                                                                                                                    79
Finegood DT, Weir GC, Bonner-Weir S. 1999. Prior streptozotocin treat-           Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir
   ment does not inhibit pancreas regeneration after 90% pancreatectomy              S, Sharpless NE. 2006. P16ink4a induces an age-dependent decline in
   in rats. Am J Physiol 276:E822-E827.                                              islet regenerative potential. Nature 443:453-457.
Freemark M, Avril I, Fleenor D, Driscoll P, Petro A, Opara E, Kendall W,         Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young
   Oden J, Bridges S, Binart N, Breant B, Kelly PA. 2002. Targeted dele-             H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D’Amour
   tion of the prl receptor: Effects on islet development, insulin production,       KA, Carpenter MK, Baetge EE. 2008. Pancreatic endoderm derived
   and glucose tolerance. Endocrinology 143:1378-1385.                               from human embryonic stem cells generates glucose-responsive insu-
Georgia S, Bhushan A. 2004. Beta cell replication is the primary mechanism           lin-secreting cells in vivo. Nat Biotechnol 26:443-452.
   for maintaining postnatal beta cell mass. J Clin Invest 114:963-968.          Lechner A, Yang YG, Blacken RA, Wang L, Nolan AL, Habener JF. 2004.
Gradwohl G, Dierich A, LeMeur M, Guillemot F. 2000. Neurogenin3 is                   No evidence for significant transdifferentiation of bone marrow into
   required for the development of the four endocrine cell lineages of the           pancreatic beta-cells in vivo. Diabetes 53:616-623.
   pancreas. Proc Natl Acad Sci U S A 97:1607-1611.                              Lee CS, De Leon DD, Kaestner KH, Stoffers DA. 2006. Regeneration of
Guz Y, Nasir I, Teitelman G. 2001. Regeneration of pancreatic beta cells             pancreatic islets after partial pancreatectomy in mice does not involve
   from intra-islet precursor cells in an experimental model of diabetes.            the reactivation of neurogenin-3. Diabetes 55:269-272.
   Endocrinology 142:4956-4968.                                                  Lenzen S. 2008. The mechanisms of alloxan- and streptozotocin-induced

                                                                                                                                                                 Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
Hanna J, Markoulaki S, Schorderet P, Carey BW, Beard C, Wernig M,                    diabetes. Diabetologia 51:216-226.
   Creyghton MP, Steine EJ, Cassady JP, Foreman R, Lengner CJ, Dausman           Li WC, Horb ME, Tosh D, Slack JM. 2005a. In vitro transdifferentiation
   JA, Jaenisch R. 2008. Direct reprogramming of terminally differentiated           of hepatoma cells into functional pancreatic cells. Mech Dev 122:835-
   mature b lymphocytes to pluripotency. Cell 133:250-264.                           847.
Hao E, Tyrberg B, Itkin-Ansari P, Lakey JR, Geron I, Monosov EZ, Barcova         Li Y, Cao X, Li LX, Brubaker PL, Edlund H, Drucker DJ. 2005b. Beta-cell
   M, Mercola M, Levine F. 2006. Beta-cell differentiation from nonendo-             pdx1 expression is essential for the glucoregulatory, proliferative, and
   crine epithelial cells of the adult human pancreas. Nat Med 12:310-               cytoprotective actions of glucagon-like peptide-1. Diabetes 54:482-
   316.                                                                              491.
Hess D, Li L, Martin M, Sakano S, Hill D, Strutt B, Thyssen S, Gray DA,          Meier JJ, Butler AE, Saisho Y, Monchamp T, Galasso R, Bhushan A, Rizza
   Bhatia M. 2003. Bone marrow-derived stem cells initiate pancreatic re-            RA, Butler PC. 2008. Beta-cell replication is the primary mechanism
   generation. Nat Biotechnol 21:763-770.                                            subserving the postnatal expansion of beta-cell mass in humans. Diabe-
Huang C, Snider F, Cross JC. 2009. Prolactin receptor is required for nor-           tes 57:1584-1594.
   mal glucose homeostasis and modulation of beta-cell mass during preg-         Minami K, Okuno M, Miyawaki K, Okumachi A, Ishizaki K, Oyama K,
   nancy. Endocrinology 150:1618-1626.                                               Kawaguchi M, Ishizuka N, Iwanaga T, Seino S. 2005. Lineage tracing
Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE,                        and characterization of insulin-secreting cells generated from adult pan-
   Melton DA. 2008a. Induction of pluripotent stem cells by defined fac-             creatic acinar cells. Proc Natl Acad Sci U S A 102:15116-15121.
   tors is greatly improved by small-molecule compounds. Nat Biotechnol          Nelson JL, Gillespie KM, Lambert NC, Stevens AM, Loubiere LS,
   26:795-797.                                                                       Rutledge JC, Leisenring WM, Erickson TD, Yan Z, Mullarkey ME,
Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S,                        Boespflug ND, Bingley PJ, Gale EA. 2007. Maternal microchimerism
   Muhlestein W, Melton DA. 2008b. Induction of pluripotent stem cells               in peripheral blood in type 1 diabetes and pancreatic islet beta cell
   from primary human fibroblasts with only oct4 and sox2. Nat Biotechnol            microchimerism. Proc Natl Acad Sci U S A 104:1637-1642.
   26:1269-1275.                                                                 Nir T, Melton DA, Dor Y. 2007. Recovery from diabetes in mice by beta cell
Hussain MA, Porras DL, Rowe MH, West JR, Song WJ, Schreiber WE,                      regeneration. J Clin Invest 117:2553-2561.
   Wondisford FE. 2006. Increased pancreatic beta-cell proliferation medi-       Okuno M, Minami K, Okumachi A, Miyawaki K, Yokoi N, Toyokuni S,
   ated by CREB binding protein gene activation. Mol Cell Biol 26:7747-              Seino S. 2007. Generation of insulin-secreting cells from pancreatic aci-
   7759.                                                                             nar cells of animal models of type 1 diabetes. Am J Physiol Endocrinol
Ianus A, Holz GG, Theise ND, Hussain MA. 2003. In vivo derivation of                 Metab 292:E158-E165.
   glucose-competent pancreatic endocrine cells from bone marrow with-           Oliver-Krasinski JM, Stoffers DA. 2008. On the origin of the beta cell.
   out evidence of cell fusion. J Clin Invest 111:843-850.                           Genes Dev 22:1998-2021.
Inada A, Nienaber C, Katsuta H, Fujitani Y, Levine J, Morita R, Sharma A,        Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, Lensch
   Bonner-Weir S. 2008. Carbonic anhydrase ii-positive pancreatic cells              MW, Cowan C, Hochedlinger K, Daley GQ. 2008. Disease-specific in-
   are progenitors for both endocrine and exocrine pancreas after birth.             duced pluripotent stem cells. Cell 134:877-886.
   Proc Natl Acad Sci U S A 105:19915-19919.                                     Patterson CC, Dahlquist GG, Gyurus E, Green A, Soltesz G. 2009. Inci-
Karnik SK, Hughes CM, Gu X, Rozenblatt-Rosen O, McLean GW, Xiong                     dence trends for childhood type 1 diabetes in Europe during 1989-2003
   Y, Meyerson M, Kim SK. 2005. Menin regulates pancreatic islet growth              and predicted new cases 2005-20: A multicentre prospective registration
   by promoting histone methylation and expression of genes encoding                 study. Lancet 373:2027-2033.
   p27kip1 and p18ink4c. Proc Natl Acad Sci U S A 102:14659-14664.               Rankin MM, Kushner JA. 2009. Adaptive beta cell proliferation is severely
Karnik SK, Chen H, McLean GW, Heit JJ, Gu X, Zhang AY, Fontaine M,                   restricted with advanced age. Diabetes 58:1365-1372.
   Yen MH, Kim SK. 2007. Menin controls growth of pancreatic beta-cells          Saisho Y, Butler AE, Manesso E, Galasso R, Toffolo GM, Cobelli C, Rizza
   in pregnant mice and promotes gestational diabetes mellitus. Science              RA, Butler PC. 2009. Beta cells are formed throughout adult life in hu-
   318:806-809.                                                                      mans primarily from sources independent of duplication of existing beta
Keller MP, Choi Y, Wang P, Belt Davis D, Rabaglia ME, Oler AT, Stapleton             cells. Diabetes 58:1582-P.
   DS, Argmann C, Schueler KL, Edwards S, Steinberg HA, Chaibub Neto             Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ,
   E, Kleinhanz R, Turner S, Hellerstein MK, Schadt EE, Yandell BS,                  Kathiresan S, Hirschhorn JN, Daly MJ, Hughes TE, Groop L, Altshuler
   Kendziorski C, Attie AD. 2008. A gene expression network model of                 D, Almgren P, Florez JC, Meyer J, Ardlie K, Bengtsson Bostrom K,
   type 2 diabetes links cell cycle regulation in islets with diabetes suscep-       Isomaa B, Lettre G, Lindblad U, Lyon HN, Melander O, Newton-Cheh
   tibility. Genome Res 18:706-716.                                                  C, Nilsson P, Orho-Melander M, Rastam L, Speliotes EK, Taskinen
Kim H, Chak E, Kishimoto N, Honig G, Yang K, Ohta Y, Wang J, Tecott L,               MR, Tuomi T, Guiducci C, Berglund A, Carlson J, Gianniny L, Hackett
   German MS. 2009. Serotonin drives pancreatic beta-cell proliferation              R, Hall L, Holmkvist J, Laurila E, Sjogren M, Sterner M, Surti A,
   druing pregnancy. Diabetes 58:1600-P.                                             Svensson M, Svensson M, Tewhey R, Blumenstiel B, Parkin M, Defelice
Kojima H, Fujimiya M, Matsumura K, Younan P, Imaeda H, Maeda M,                      M, Barry R, Brodeur W, Camarata J, Chia N, Fava M, Gibbons J,
   Chan L. 2003. NeuroD-betacellulin gene therapy induces islet neogen-              Handsaker B, Healy C, Nguyen K, Gates C, Sougnez C, Gage D, Nizzari
   esis in the liver and reverses diabetes in mice. Nat Med 9:596-603.               M, Gabriel SB, Chirn GW, Ma Q, Parikh H, Richardson D, Ricke

80                                                                                                                                               ILAR Journal
D, Purcell S. 2007. Genome-wide association analysis identifies loci for      Vasavada RC, Garcia-Ocana A, Zawalich WS, Sorenson RL, Dann P, Syed
    type 2 diabetes and triglyceride levels. Science 316:1331-1336.                   M, Ogren L, Talamantes F, Stewart AF. 2000. Targeted expression of
Schraenen A, Lemaire K, Hendrickx N, Gravnik M, Van Lommel L, Schuit                  placental lactogen in the beta cells of transgenic mice results in beta cell
    FC. 2009. Serotonin competence of pancreatic beta cells during preg-              proliferation, islet mass augmentation, and hypoglycemia. J Biol Chem
    nancy. Diabetes 58:1631-P.                                                        275:15399-15406.
Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos             Voltarelli JC, Couri CE, Stracieri AB, Oliveira MC, Moraes DA, Pieroni F,
    MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L,                      Barros GM, Madeira MI, Malmegrim KC, Foss-Freitas MC, Simoes BP,
    Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely               Foss MC, Squiers E, Burt RK. 2008. Autologous hematopoietic stem cell
    KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart                   transplantation for type 1 diabetes. Ann N Y Acad Sci 1150:220-229.
    MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan           Wang S, Jensen JN, Seymour PA, Hsu W, Dor Y, Sander M, Magnuson MA,
    TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW,                      Serup P, Gu G. 2009. Sustained neurog3 expression in hormone-
    Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M. 2007.                 expressing islet cells is required for endocrine maturation and function.
    A genome-wide association study of type 2 diabetes in Finns detects               Proc Natl Acad Sci U S A 106:9715-9720.
    multiple susceptibility variants. Science 316:1341-1345.                      Wang X, Ge S, Gonzalez I, McNamara G, Rountree CB, Xi KK, Huang G,
Song WJ, Schreiber WE, Zhong E, Liu FF, Kornfeld BD, Wondisford FE,                   Bhushan A, Crooks GM. 2006. Formation of pancreatic duct epithelium
    Hussain MA. 2008. Exendin-4 stimulation of cyclin a2 in beta-cell pro-            from bone marrow during neonatal development. Stem Cells 24:307-314.

                                                                                                                                                                     Downloaded from https://academic.oup.com/ilarjournal/article/51/1/74/720461 by guest on 04 January 2021
    liferation. Diabetes 57:2371-2381.                                            Wang ZV, Mu J, Schraw TD, Gautron L, Elmquist JK, Zhang BB, Brownlee
Spence JR, Wells JM. 2007. Translational embryology: Using embryonic                  M, Scherer PE. 2008. Panic-attac: A mouse model for inducible and
    principles to generate pancreatic endocrine cells from embryonic stem             reversible beta-cell ablation. Diabetes 57:2137-2148.
    cells. Dev Dyn 236:3218-3227.                                                 Xu G, Stoffers DA, Habener JF, Bonner-Weir S. 1999. Exendin-4 stimulates
Stadtfeld M, Brennand K, Hochedlinger K. 2008a. Reprogramming of pan-                 both beta-cell replication and neogenesis, resulting in increased beta-
    creatic beta cells into induced pluripotent stem cells. Curr Biol 18:890-         cell mass and improved glucose tolerance in diabetic rats. Diabetes
    894.                                                                              48:2270-2276.
Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. 2008b. Induced           Xu X, D’Hoker J, Stange G, Bonne S, De Leu N, Xiao X, Van de Casteele
    pluripotent stem cells generated without viral integration. Science               M, Mellitzer G, Ling Z, Pipeleers D, Bouwens L, Scharfmann R, Gradwohl
    322:945-949.                                                                      G, Heimberg H. 2008. Beta cells can be generated from endogenous
Stoffers DA, Kieffer TJ, Hussain MA, Drucker DJ, Bonner-Weir S, Habener               progenitors in injured adult mouse pancreas. Cell 132:197-207.
    JF, Egan JM. 2000. Insulinotropic glucagon-like peptide 1 agonists            Yamashita H, Shao J, Ishizuka T, Klepcyk PJ, Muhlenkamp P, Qiao L,
    stimulate expression of homeodomain protein idx-1 and increase islet              Hoggard N, Friedman JE. 2001. Leptin administration prevents sponta-
    size in mouse pancreas. Diabetes 49:741-748.                                      neous gestational diabetes in heterozygous lepr(db/+) mice: Effects on
Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from               placental leptin and fetal growth. Endocrinology 142:2888-2897.
    mouse embryonic and adult fibroblast cultures by defined factors. Cell        Yechoor V, Liu V, Espiritu C, Paul A, Oka K, Kojima H, Chan L. 2009.
    126:663-676.                                                                      Neurogenin3 is sufficient for transdetermination of hepatic progenitor
Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K,                      cells into neo-islets in vivo but not transdifferentiation of hepatocytes.
    Yamanaka S. 2007. Induction of pluripotent stem cells from adult                  Dev Cell 16:358-373.
    human fibroblasts by defined factors. Cell 131:861-872.                       Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin, II, Thomson JA.
Teta M, Rankin MM, Long SY, Stein GM, Kushner JA. 2007. Growth and                    2009. Human induced pluripotent stem cells free of vector and trans-
    regeneration of adult beta cells does not involve specialized progenitors.        gene sequences. Science 324:797-801.
    Dev Cell 12:817-826.                                                          Zhang D, Jiang W, Liu M, Sui X, Yin X, Chen S, Shi Y, Deng H. 2009.
Thowfeequ S, Li WC, Slack JM, Tosh D. 2009. Reprogramming of liver to                 Highly efficient differentiation of human ES cells and iPS cells into
    pancreas. Methods Mol Biol 482:407-418.                                           mature pancreatic insulin-producing cells. Cell Res 19:429-438.
Tschen SI, Dhawan S, Gurlo T, Bhushan A. 2009. Age-dependent decline in           Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. 2008. In vivo repro-
    beta cell proliferation restricts the capacity of beta cell regeneration in       gramming of adult pancreatic exocrine cells to beta-cells. Nature
    mice. Diabetes 58:1312-1320.                                                      455:627-632.

Volume 51, Number 1             2010                                                                                                                          81
You can also read