With cell therapies based on potentially autologous nonpancreatic cells, targeted by gene expression vectors in vivo, or retrieved surgically, engineered ex vivo, and returned to the ho
Trang 1I Introduction
II Cell Types for Pancreatic Substitutes
III Construct Technology
IV In Vivo Implantation
V Concluding Remarks
VI Acknowledgments VII References
Bioartifi cial Pancreas
Athanassios Sambanis
Principles of Tissue Engineering, 3 rd Edition
ed by Lanza, Langer, and Vacanti
Copyright © 2007, Elsevier, Inc.
All rights reserved.
I INTRODUCTION
Diabetes is a signifi cant health problem, affecting an
estimated 20.8 million people in the United States alone,
with nearly 1.8 million affl icted with type 1 diabetes [http://
diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm#7]
Type 1 diabetes results from the loss of insulin-producing
cell mass (the β-cells of pancreatic islets) due to
autoim-mune attack Type 2 diabetes has a more complicated
disease etiology and can be the result of not producing
enough insulin and/or the body’s developing a resistance to
insulin Although initially controlled by diet, exercise, and
oral medication, type 2 diabetes often progresses toward
insulin dependence It is estimated that insulin-dependent
diabetics (both types 1 and 2) exceed 4 million people in the
United States Although insulin-dependent diabetes (IDD)
is considered a chronic disease, even the most vigilant
insulin therapy cannot reproduce the precise metabolic
control present in the nondiseased state The poor temporal
match between glucose load and insulin activity leads to a
number of complications, including increased risk of heart
disease, kidney failure, blindness, and amputation due to
peripheral nerve damage Providing more physiological
control would alleviate many of the diabetes-related health
problems, as suggested by fi ndings from the Diabetes
Control and Complications Trial (The Diabetes Control and
Complications Trial Research Group, 1997) and its
continu-ation study (DCCT/EDIC NEJM 353(25):2643–53, 2005)
Cell-based therapies, which provide continuous regulation
of blood glucose through physiologic secretion of insulin, have the potential to revolutionize diabetes care
Several directions are being considered for cell-based therapies of IDD, including implantation of immunopro-tected allogeneic or xenogeneic islets, of continuous cell lines, or of engineered non-β-cells For allogeneic islet trans-plantation, a protocol developed by physicians at the Uni-
versity of Edmonton (Shapiro et al., 2001a, 2001b, 2001c,
Bigam and Shapiro, 2004) has dramatically improved the survivability of grafts The protocol uses human islets from cadaveric donors, which are implanted in the liver of care-fully selected diabetic recipients via portal vein injection The success of the Edmonton protocol is attributed to two modifi cations relative to earlier islet transplantation studies: the use of a higher number of islets and the implementation
of a more benign, steroid-free immunosuppressive regimen However, two barriers prevent the widespread application
of this therapy The fi rst is the limited availability of human tissue, because generally more than one cadaveric donor pancreas is needed for the treatment of a single recipient The second is the need for life-long immunosuppression, which, even with the more benign protocols, results in long-term side effects to the patients
A tissue-engineered pancreatic substitute aims to address these limitations by using alternative cell sources, relaxing the cell availability limitation, and by reducing or eliminating the immunosuppressive regimen necessary for survival of the graft A number of signifi cant challenges are
Trang 2facing the development of such a substitute, however These
include procuring cells at clinically relevant quantities; the
immune acceptance of the cells, which is exacerbated in
type 1 diabetes by the resident autoimmunity in the patients;
and the fact that diabetes is not an immediately
life-threat-ening disease, so any other therapy will have to be more
effi cacious and/or less invasive than the current standard
treatment of daily blood glucose monitoring and insulin
injections
In general, developing a functional living-tissue
replace-ment requires advances and integration of several types of
technology (Nerem and Sambanis, 1995) These are (1) cell
technology, which addresses the procurement of functional
cells at the levels needed for clinical applications; (2)
con-struct technology, which involves combining the cells with
biomaterials in functional three-dimensional confi
gura-tions Construct manufacturing at the appropriate scale,
and preservation, as needed for off-the-shelf availability,
also fall under this set of technologies; (3) technologies for
in vivo integration, which address the issues of construct
immune acceptance, in vivo safety and effi cacy, and
moni-toring of construct integrity and function postimplantation
The same three types of technology need also be developed
for a pancreatic substitute It should be noted, however, that
the critical technologies differ, depending on the type of
cells used With allogeneic or xenogeneic islets or beta-cells,
the major challenge is the immune acceptance of the
implant In this case, encapsulation of the cells in
permse-lective membranes, which allow passage of
low-molecular-weight nutrients and metabolites but exclude larger
antibodies and cytotoxic cells of the host, may assist the
immune acceptance of the graft With cell therapies based
on potentially autologous nonpancreatic cells, targeted
by gene expression vectors in vivo, or retrieved surgically,
engineered ex vivo, and returned to the host, the major
challenge is engineering insulin secretion in precise response
to physiologic stimuli Lastly, with stem or progenitor cells, the primary hurdle is their reproducible differentiation into cells of the pancreatic β-phenotype Figure 42.1 shows schematically the two general therapeutic approaches based
on allo- or xenogeneic cells (Fig 42.1A) or autologous cells (Fig 42.1B)
This chapter is therefore organized as follows We fi rst describe the types of cells that have been used or are of potential use in engineering a pancreatic substitute We then discuss issues of construct technology, specifi cally encapsulation methods and the relevant biomaterials, man-ufacturing issues, and preservation of the constructs The
challenges of in vivo integration and results from in vivo
experiments with pancreatic substitutes are presented next
We conclude by offering a perspective on the current status and the future challenges in developing an effi cacious, clini-cally applicable bioartifi cial pancreas
II CELL TYPES FOR PANCREATIC SUBSTITUTES
Islets
Despite several efforts, the in vitro expansion of primary
human islets has met with limited success Adult human islets are diffi cult to propagate in culture, and their expan-sion leads to dedifferentiation, generally manifested as loss
of insulin secretory capacity Although there exist reports on
the redifferentiation of expanded islets (Lechner et al., 2005;
Ouziel-Yahalom et al., 2006) and of nonislet pancreatic cells, which are discarded after islet isolation (Todorov et al., 2006), the phenotypic stability and the in vivo effi cacy of
these cells remain unclear Additionally, with expanded and
A Allo- or Xenogeneic Cells B Autologous Cells
Implantation
Cell retrieval
Ex vivo
manipulation
Cell storage
Cell storage
Cell amplification
Cell encapsulation
Implantation
Capsule storage
Ex vivo
manipulation
Cell storage
Cell storage
Cell amplification
Cell encapsulation
Implantation
Capsule storage Capsule storage
Cell
gene therapy
FIG 42.1 Approaches for bioartifi cial pancreas development using allo- or xenogeneic cells (A) and autologous cells (B) In (A), islets are procured from
pancreatic tissue, or cell lines are thawed from cryostorage and expanded in culture; cells are encapsulated for immunoprotection before they are implanted
to achieve a therapeutic effect; encapsulated cells may also be cryopreserved for inventory management and sterility testing In (B), cells are retrieved
surgi-cally from the patient; manipulated ex vivo phenotypisurgi-cally and/or genetisurgi-cally in order to express β-cell characteristics, and in particular physiologically
responsive insulin secretion; the cells are implanted for a therapeutic effect either by themselves or, preferably, after incorporation in a three-dimensional
substitute; some of the cells may be cryopreserved for later use by the same individual In in vivo gene therapy approaches, a transgene for insulin
expres-sion is directly introduced into the host and expressed by cells in nonpancreatic tissues
Trang 3I I C E L L T Y P E S F O R P A N C R E A T I C S U B S T I T U T E S • 621
redifferentiated islets, it remains unknown whether the
insulin-secreting cells arose from the redifferentiation of
mature endocrine cells or from an indigenous stem or
pro-genitor cell population in the tissue isolate (Todorov et al.,
2006)
Animal, such as porcine, islets are amply available, and
porcine insulin is very similar to human, differing by only
one amino acid residue However, the potential use of
porcine tissue is hampered by the unlikely but possible
transmission of porcine endogenous retroviruses (PERV) to
human hosts as well as by the strong xenograft
immuno-genicity that they elicit Use of closed, PERV-free herds is
reasonably expected to alleviate the fi rst problem With
regard to immunogenicity, a combination of less
immuno-genic islets, islet encapsulation in permselective barriers,
and host immunosuppression may yield long-term survival
of the implant The use of transgenic pigs that do not express
the α-Gal (α[1,3]-galactose) epitope is one possible approach
for reducing the immunogenicity of the islets Studies also
indicate that neonatal pig islets induce a lower T-cell
reac-tivity than adult islets (Bloch et al., 1999), even though the
α-Gal epitope is abundant in neonatal islets as well (Rayat
et al., 1998) Furthermore, it is possible that the primary
antigenic components in islet tissue are the ductal epithelial
and vascular endothelial cells, which express prominently
the α-Gal epitope; on the other hand, β-cells express the
epitope immediately after isolation but not after
mainte-nance in culture (Heald et al., 1999) It should also be noted
that the large-scale isolation of porcine islets under
condi-tions of purity and sterility that will be needed for eventual
regulatory approval pose some major technical hurdles,
which have not been addressed yet
-Cell Lines
Recognizing the substantial diffi culties involved with
the procurement and amplifi cation of pancreatic islets,
several investigators have pursued the development of
con-tinuous cell lines, which can be amplifi ed in culture yet
retain key differentiated properties of normal β-cells One of
the fi rst successful developments in this area was the
gen-eration of the βTC family of insulinomas, derived from
transgenic mice carrying a hybrid insulin-promoted simian
virus 40 tumor antigen gene; these cells retained their
dif-ferentiated features for about 50 passages in culture (Efrat
et al., 1988) The hypersensitive glucose responsiveness of
the initial βTC lines was reportedly corrected in subsequent
lines by ensuring expression of glucokinase and of the
high-K m glucose transporter Glut2, and with no or low expression
of hexokinase and of the low-K m transporter Glut1 (Efrat et
al., 1993; Knaack et al., 1994) A similar approach was used
to develop the mouse MIN-6 cell line that exhibits
glucose-responsive secretion of endogenous insulin (Miyazaki et al.,
1990) Subsequently, Efrat and coworkers developed the
βTC-tet cell line, in which expression of the SV40 T antigen
(Tag) oncoprotein is tightly and reversibly regulated by
tet-racycline Thus, cells proliferate when Tag is expressed, and
shutting off Tag expression halts cell growth (Efrat et al.,
1995; Efrat, 1998) Such reversible transformation is an elegant approach in generating a supply of β-cells via pro-liferation of an inoculum, followed by arrest of the growth
of cells when the desirable population size is reached When retained in capsules, proliferating cells do not grow uncon-trollably, since the dissolved-oxygen concentration in the surrounding milieu can support up to a certain number of viable, metabolically active cells in the capsule volume This number of viable cells is maintained through equilibration
of cell growth and death processes (Papas et al., 1999a,
1999b) Thus, growth arrest is useful primarily in preventing the growth of cells that have escaped from broken capsules
in vivo and in reducing the cellular turnover in the capsules
The latter reduces the number of accumulated dead cells in the implant and thus the antigenic load to the host affected
by proteins from dead and lysed cells that pass through the capsule material
In a different approach, Newgard and coworkers (Clark
et al., 1997) carried out a stepwise introduction of genes
related to β-cell performance into a poorly secreting rat insulinoma (RIN) line In particular, RIN cells were itera-tively engineered to stably express multiple copies of the insulin gene, the glucose transporter Glut2, and the gluco-kinase gene, which are deemed essential for proper expres-sion of β-cell function Although this is an interesting methodology, it is doubtful that all genes necessary for reproducing β-cell function can be identifi ed and stably expressed in a host cell Recently, signifi cant progress was made toward establishing a human pancreatic β-cell line that appears functionally equivalent to normal β-cells
(Narushima et al., 2005) This was accomplished through a
complicated procedure involving retroviral transfection of primary β cells with the SV40 large T antigen and cDNAs of human telomerase reverse transcriptase This resulted in a reversibly immortalized human β-cell clone, which secreted insulin in response to glucose, expressed β-cell transcrip-tional factors, prohormone convertases 1/3 and 2, which process proinsulin to mature insulin, and restored normo-glycemia upon implantation in diabetic immunodefi cient
mice (Narushima et al., 2005).
With regard to β-cell lines capable of proliferation under the appropriate conditions, key issues that remain to be addressed include (1) their long-term phenotypic stability,
in vitro and in vivo; (2) their potential tumorigenicity, if cells
escape from an encapsulation device, especially when these cells are allografts that may evade the hosts’ immune defenses for a longer period of time than acutely rejected xenografts; and (3) their possible recognition by the auto-immune rejection mechanism in type 1 diabetic hosts
Engineered Non– Pancreatic Cells
The use of non–β pancreatic cells from the same patient, engineered for insulin secretion, relaxes both the cell availability and immune acceptance limitations that exist with other types of cells It has been shown that the A-chain/
Trang 4C-peptide and B-chain/C-peptide cleavage sites on the
pro-insulin gene can be mutated so that the ubiquitous
endo-peptidase furin recognizes and completely processes
proinsulin into mature insulin absent of any intermediates
(Yanagita et al., 1992) Based on this concept, several
non-endocrine cell lines have been successfully transfected to
produce immunoreactive insulin, including hepatocytes,
myoblasts, and fi broblasts (Yanagita et al., 1993) In a
differ-ent approach, Lee and coworkers (2000) expressed a
syn-thetic single-chain insulin analog, which does not require
posttranslational processing, in hepatocytes Although
recombinant insulin expression is relatively straightforward,
a key remaining challenge is achieving the tight regulation
of insulin secretion in response to physiologic stimuli, which
is needed for achieving normoglycemia in higher animals
and, eventually, humans
One approach for achieving regulation of insulin
secre-tion is through regulasecre-tion of biosynthesis at the gene
tran-scription level, as realized in hepatocytes by Thule et al
(Thule et al., 2000; Thule and Liu, 2000) and Lee et al (2000)
Besides the ability to confer transcriptional-level regulation,
hepatocytes are particularly attractive as producers of
recombinant insulin due to their high synthetic and
secre-tory capacity and their expression of glucokinase and Glut2
(Cha et al., 2000; Lannoy et al., 2002) Hepatic delivery by
viral vectors and expression of the glucose-responsive
insulin transgene in diabetic rats controlled the
hypergly-cemic state for extended periods of time (Lee et al., 2000;
Thule and Liu, 2000; Olson et al., 2003) Nevertheless,
trans-criptional regulation is sluggish, involving long time lags
between stimulation of cells with a secretagogue and
induced insulin secretion as well as between removal of the
secretagogue and down-regulation of the secretory response
(Tang and Sambanis, 2003) The latter is physiologically
more important, because it means that the cells continue to
secrete insulin after glucose has been down-regulated, thus
resulting in potentially serious hypoglycemic excursions
Increasing the number of stimulatory glucose elements in a
promoter enhances the cellular metabolic responsiveness
in vitro (Thule et al., 2000) With regard to secretion
down-regulation, Tang and Sambanis (2003) hypothesized that the
slow kinetics of this process following removal of the
tran-scriptional activator are due to the stability of the
preproin-sulin mRNA, which continues to become translated after
transcription has been turned off Using a modifi ed
prepro-insulin cDNA that produced an mRNA with two more copies
of the insulin gene downstream of the stop codon resulted
in preproinsulin mRNA subjected to nonsense mediated
decay and thus destabilized This signifi cantly expedited the
kinetics of secretion down-regulation on turning off
tran-scription (Tang and Sambanis, 2003) Thus, the
combina-tion of optimal transcripcombina-tional regulacombina-tion with transgene
message destabilization promises further improvements in
insulin secretion dynamics from transcriptionally regulated
hepatic cells It should be noted, however, that despite the
time delays inherent in transcriptional regulation, hepatic insulin gene therapy is suffi cient to sustain vascular nitric oxide production and inhibit acute development of diabetes-associated endothelial dysfunction in diabetic
rats (Thule et al., 2006) Hence, many aspects of the
thera-peutic potential of hepatic insulin expression remain to be explored
Another appealing target cell type is endocrine cells, which possess a regulated secretory pathway and the enzymatic machinery needed to process authentic proinsu-lin into insulin Early work in this area involved expression
of recombinant insulin in the anterior pituitary mouse
AtT20 cell line (Moore et al., 1983), which can be sub
-jected to repeated episodes of induced insulin secretion
using nonmetabolic secretagogues (Sambanis et al., 1990)
Cotransfection with genes encoding the glucose transporter Glut-2 and glucokinase resulted in glucose-responsive
insulin secretion (Hughes et al., 1992, 1993) However,
limi-tations of this approach include possible instabilities in the cellular phenotype and the continued secretion of endoge-nous hormones, such as adrenocorticotropic hormone from AtT-20 cells, which are not compatible with prandial metabolism
In this regard, endocrine cells of the intestinal lium, or enteroendocrine cells, are especially promising
epithe-Enteroendocrine cells secrete their incretin products in a tightly controlled manner that closely parallels the secretion
of insulin following oral glucose load in human subjects;
incretin hormones are fully compatible with prandial
metabolism and glucose regulation (Schirra et al., 1996;
Kieffer and Habener, 1999) As with β-cells, enteroendocrine cells are polar, with sensing microvilli on their luminal side and secretory granules docked at the basolateral side, adja-cent to capillaries Released incretin hormones include the glucagon-like peptides (GLP-1 and GLP-2) from intestinal L-cells and glucose-dependent insulinotropic polypeptide (GIP) from K-cells, which potentiate insulin production from the pancreas after a meal (Drucker, 2002) The impor-tance of enteroendocrine cells (and, in particular, L-cells) was fi rst put forward by Creutzfeldt (1974), whose primary interest in these cells was for the prospect of using GLP-1 for the treatment of type 2 diabetes Furthermore, ground-
breaking work by Cheung et al (2000) demonstrated that
insulin produced and secreted by genetically modifi ed intestinal K-cells of transgenic mice prevented the animals from becoming diabetic after injection with streptozotocin (STZ), which specifi cally kills the β-cells of the pancreas
This is an important proof-of-concept study, which showed that enteroendocrine cell–produced insulin can provide regulation of blood glucose levels Subsequent work with a human intestinal L-cell line demonstrated that these cells can be effectively transduced to express recombinant human insulin, which colocalizes in secretory vesicles with endog-enous GLP-1 and thus is secreted with identical kinetics to GLP-1 in response to stimuli (Tang and Sambanis, 2003,
Trang 52004) The intestinal tract could be considered an attractive
target for gene therapy because of its large size, making it
the largest endocrine organ in the body (Wang et al., 2004);
however, enteroendocrine cell gene therapy faces serious
diffi culties due to anatomic complexity, with the
entero-endocrine cells being located at the base of invaginations of
the gut mucosa called crypts, the very harsh conditions in
the stomach and intestine, and the rapid turnover of the
intestinal epithelium
Contrary to direct in vivo gene delivery, ex vivo
gene therapy involves retrieving the target cells surgically,
culturing them and possibly expanding them in vitro,
genetically engineering them to express the desired
proper-ties, and then returning them to the host, either as such or
in a three-dimensional tissue substitute It is generally
thought that the ex vivo approach is advantageous, for
it allows for the thorough characterization of the genetically
engineered cells prior to implantation, possibly for the
preservation of some of the cells for later use by the
same individual and, importantly, for localization and
retrievability of the implant However, the challenges
imposed by the ex vivo approach, including the surgical
retrieval, culturing, and in vitro genetic engineering of the
target cells are signifi cant, so such methods are currently
under development
Differentiated Stem or Progenitor Cells
Naturally, throughout life, islets turn over slowly, and
new, small islets are continually generated from ductal
pro-genitors (Finegood et al., 1995; Bonner-Weir and Sharma,
2002) There is also considerable evidence that adult
plu-ripotent stem cells may be a possible source of new islets
(Bonner-Weir et al., 2000; Ramiya et al., 2000; Kojima et al.,
2003) However, efforts to regenerate β-cells in vitro or in
vivo by differentiation of embryonic or adult stem or
pan-creatic progenitor cells have produced mixed results
Insulin-producing, glucose-responsive cells, as well as other
pan-creatic endocrine cells, have been generated from mouse
embryonic stem cells (Lumelsky et al., 2001)
Insulin-secret-ing cells obtained from embryonic stem cells reversed
hyperglycemia when implanted in mice rendered diabetic
by STZ injection (Soria et al., 2000) In another study, mouse
embryonic stem cells transfected to express constitutively
Pax4, a transcription factor essential for β-cell development,
differentiated into insulin-producing cells and normalized
blood glucose when implanted in STZ-diabetic mice
(Blyszczuk et al., 2003) On the other hand, other studies do
not support differentiation of embryonic stem cells into the
β-cell phenotype (Rajagopal et al., 2003) Overall, the mixed
and somewhat inconsistent results point to the
consider-able work that needs to be done before stem or progenitor
cells can be reliably differentiated into β cells at a clinically
relevant scale Harnessing the in vivo regenerative capacity
of the pancreatic endocrine system may present a
promis-ing alternative approach
Engineering of Cells for Enhanced
Survival In Vivo
Because islets and other insulin-secreting cells
experi-ence stressful conditions during in vitro handling and in
vivo postimplantation, several strategies have been
imple-mented to enhance islet or nonislet cell survival in atic substitutes Strategies generally focus on improving the immune acceptance of the graft, enhancing its resistance to cytokines, and reducing its susceptibility to apoptosis Phe-notypic manipulations include extended culturing of neo-natal and pig islets at 37°C, which apparently reduces their immunogenicity, possibly by down-regulating the major histocompatibility class 1 antigens on the islet surface; islet pretreatment with TGF-β1; and enzymatic treatment of pig islets with α-galactosidase to reduce the a-galactosyl epitope
pancre-on islets (Prokop, 2001) However, the permanency of these modifi cations is unknown For instance, a-galactosyl epi-topes reappear on islets 48 hours after treatment with α-galactosidase With proliferative cell lines destined for recombinant insulin expression, selection of clones resis-
tant to cytokines appears feasible (Chen et al., 2000) Gene
chip analysis of resistant cells may then be used to identify the genes responsible for conferring cytokine resistance
Genetic modifi cations for improving survival in vivo
may offer prolonged expression of the desired properties relative to phenotypic manipulations, but they also present the possibility of modifying the islets in additional, undesir-able ways Notable among the various proposed approaches, reviewed in Jun and Yoon (2005), are the expression of the immunomodulating cytokines IL-4 or a combination of IL-
10 and TGF-β, which promoted graft survival by preventing immune attack in mice; and the expression of the antiapop-totic bcl-2 gene using a replication defective herpes simplex virus, which resulted in protection of β-cells from a cytokine mixture of interleukin-1β, TNF-α, and IFN-γ in vitro.
III CONSTRUCT TECHNOLOGY
Construct technology focuses on associating cells with biocompatible materials in functional three-dimensional confi gurations Depending on the type of cells used, the primary function of the construct can be one or more of the following: to immunoprotect the cells postimplantation, to
enable cell function, to localize insulin delivery in vivo, or
to provide retrievability of the implanted cells
Encapsulated Cell Systems
Encapsulation for immunoprotection involves rounding the cells with a permselective barrier, in essence
sur-an ultrafi ltration membrsur-ane, which allows passage of molecular-weight nutrients and metabolites, including insulin, but excludes larger antibodies and cytototoxic cells
low-of the host Figure 42.2 summarizes the common types low-of encapsulation devices, which include spherical microcap-sules, tubular or planar diffusion chambers, thin sheets, and vascular devices
I I I C O N S T R U C T T E C H N O L O G Y • 623
Trang 6Encapsulation can be pursued via one of two general
approaches With capsules fabricated using water-based
chemistry, cells are fi rst suspended in un-cross-linked
polymer, which is then extruded as droplets into a solution
of the cross-linking agent A typical example here is the very
commonly used alginate encapsulation Alginate is a
complex mixture of polysaccharides obtained from
sea-weeds, which forms a viscous solution in physiologic saline
Islets or other insulin-secreting cells are suspended in
sodium alginate, and droplets are extruded into a solution
of calcium chloride Calcium cross-links alginate,
instanta-neously trapping the cells within the gel The size of the
droplets, hence also of the cross-linked beads, can be
con-trolled by fl owing air parallel to the extrusion needle so that
droplets detach at a smaller size than if they were allowed
to fall by gravity; or by using an electrostatic droplet
genera-tor, in which droplets are detached from the needle by
adjusting the electrostatic potential between the needle and
the calcium chloride bath Capsules generated this way can
have diameters from a few hundred micrometers to more
than one millimeter Alginate by itself is relatively
perme-able; to generate the permselective barrier, beads are treated
with a polycationic solution, such as l-lysine or
poly-l-ornithine The reaction time between alginate and the
polycation determines the molecular weight cutoff of the
resulting membrane Poly-l-lysine is highly infl ammatory
in vivo, however, so beads are coated with a fi nal layer of
alginate to improve their biocompatibility Hence, calcium
alginate/poly-l-lysine/alginate (APA) beads are fi nally
formed Treating the beads with a calcium chelator, such as
citrate, presumably liquefi es the inner core, forming APA
membranes Other materials that have been used for cell
microencapsulation include agarose, photo-cross-linked
poly(ethylene glycol), and (ethyl methacrylate, methyl
methacrylate, and dimethylaminoethyl methacrylate)
copolymers (Mikos et al., 1994; Sefton and Kharlip, 1994)
Advantages of hydrogel microcapsules include a high
surface-to-volume ratio, and thus good transport
proper-ties, as well as ease of handling and implantation Small beads can be implanted in the peritoneal cavity of animals simply by injection, without the need for incision Other common implantation sites include the subcutaneous space and the kidney capsules Disadvantages include the fragility
of the beads, especially if the cross-linking cation becomes chelated by compounds present in the surrounding milieu
or released by lysed cells, and the lack of easy retrievability once the beads have been dispersed in the peritoneal cavity
of a host Earlier problems caused by the variable tion of alginates and the presence of endotoxins have been resolved through the development and commercial avail-ability of ultrapure alginates of well-defi ned molecular
composi-weight and composition (Sambanis, 2000; Stabler et al.,
2001)
Hydrogels impose little diffusional resistance to solutes, and indeed effective diffusivities in calcium alginate and agarose hydrogels are in the range of 50–100% of the corre-sponding diffusivities in water (Tziampazis and Sambanis, 1995; Lundberg and Kuchel, 1997) However, with conven-tional microencapsulation, the volume of the hydrogel con-tributes signifi cantly to the total volume of the implant For example, with a 500-µm microcapsule containing a 300-µm islet, the polymer volume constitutes 78% of the total capsule volume Additionally, conventional microcapsules may not
be appropriate for hepatic portal vein implantation because, besides their higher implant volume relative to the same number of naked islets, they may result in higher portal vein pressure and more incidences of blood coagulation in the liver To address this problem, methods have been devel-oped for islet encapsulation in thin conformal polymeric coats Materials that have been used for conformal coating include photopolymerized poly(ethylene glycol) diacrylate
(Hill et al., 1997; Cruise et al., 1999) and hydroxyethyl
methacrylate-methyl methacrylate (HEMA-MMA) (May
and Sefton, 1999; Sefton et al., 2000).
Encapsulated cell systems can also be fabricated by forming the permselective membrane in a tubular or disc-
pre-Vascular device
Microcapsules
Membrane chambers
Titanium housing
Immunobarrier membranes
Silicone rubber spacer
Immunobarrier membranes
Silicone rubber spacer
Trang 7shaped confi guration, fi lling the construct with a suspension
of islets or other insulin-secreting cells in an appropriate
extracellular matrix, and then sealing the device This
approach is particularly useful when organic solvents or
other chemicals harsh to the cells are needed for the
fabrica-tion of the membranes Membrane chambers can be of
tubular or planar geometry (Fig 42.2) The cells are
sur-rounded by the semipermeable membrane and can be
implanted intraperitoneally, subcutaneously, or at other
sites Membrane materials used in fabricating these devices
include polyacrylonitrile-polyvinyl chloride (PAN-PVC)
copolymers, polypropylene, polycarbonate, cellulose nitrate,
and polyacrylonitrile-sodium methallylsulfonate (AN69)
(Lanza et al., 1992; Mikos et al., 1994; Prevost et al., 1997;
Delaunay et al., 1998; Sambanis, 2000) Typical values of
device thickness or fi ber diameter are 0.5–1 mm
Advan-tages of membrane chambers are the relative ease of
han-dling, the fl exibility with regard to the matrix in which the
cells are embedded, and retrievability after implantation A
major disadvantage is their inferior transport properties,
since the surface-to-volume ratio is smaller than that of
microcapsules and diffusional distances are longer
Constructs connected to the vasculature via an
arterio-venus shunt consist of a semipermeable tube
sur-rounded by the cell compartment (Fig 42.2) The tube
is connected to the vasculature, and transport of solutes
between the blood and the cell compartment occurs via
the pores in the tube wall A distinct advantage of the
vascular device is the improved transport of nutrients
and metabolites, which occurs by both diffusion and
con-vection However, the major surgery that is needed for
implantation and problems of blood coagulation at the
anastomosis sites have considerably reduced enthusiasm
for these devices
Other Construct Systems
A common approach for improving the oxygenation of
cells in diffusion chambers is to encourage the formation of
neovasculature around the implant This is discussed in the
following “In Vivo Implantation” section Other innovative
approaches that have been proposed include the
electro-chemical generation of oxygen in a device adjacent to a
planar immunobarrier diffusion chamber containing the
insulin-secreting cells (Wu et al., 1999); and the
coencapsu-lation of islets with algae, where the latter produce oxygen
photosynthetically upon illumination (Bloch et al., 2006)
These were in vitro studies, however, and the ability to
translate these approaches to effective in vivo confi
gura-tions remains unknown
In a different design, Cheng et al (2004, 2006)
combined constitutive insulin-secreting cells with a
glucose-responsive material in a disc-shaped construct As indicated
earlier, it is straightforward to genetically engineer
non-β-cells for constitutive insulin secretion; the challenge is
in engineering appropriate cellular responsiveness to
physiologic stimuli In this proposed device, a concanavalin
A (con A)–glycogen material, sandwiched between two ultrafi ltration membranes, acted as a control barrier to insulin release from an adjacent compartment containing the cells Specifi cally, con A–glycogen formed a gel at a low concentration of glucose, which was reversibly con-verted to sol at a high glucose concentration, as glucose displaced glycogen from the gel network Since insulin dif-fusivity is higher through the sol than through the gel, insu-lin released by the cells during low-glucose periods diffused slowly through the gel material; when switched
to high glucose, the insulin accumulated in the cell compartment during the previous cycle was released at a faster rate through the sol-state polymer Overall, this approach converted the constitutive secretion of insulin
by the cells to a glucose-responsive insulin release by the
device (Cheng et al., 2006) Again, these were in vitro studies, and the in vivo effi cacy of this approach remains to be
evaluated
Construct Design and In Vitro Evaluation
Design of three-dimensional encapsulated systems can
be signifi cantly enabled using mathematical models of solute transport through the tissue and of nutrient consumption and metabolite production by the cells Beyond the microvasculature surrounding the construct, transport of solutes occurs by diffusion, unless the construct
is placed in a fl ow environment, in which case convective transport may also occur Due to its low solubility, transport
of oxygen to the cells is the critical issue Models can be used
to evaluate the dimensions and the cell density within the construct so that all cells are suffi ciently nourished and the capsule as a whole is rapidly responsive to changes in the surrounding glucose concentration (Tziampazis and Sambanis, 1995) Experimental and modeling methods for determining transport properties and reaction kinetics have been described previously (Sambanis and Tan, 1999) Furthermore, models can be developed to account for the cellular reorganization that occurs in constructs with time
as a result of cell growth, death, and possibly migration processes Such reorganization is especially signifi cant when proliferating insulin-secreting cell lines are encapsu-
lated in hydrogel matrices (Stabler et al., 2001; Simpson
et al., 2005; Gross et al., 2007).
Pancreatic tissue substitutes should be evaluated in
vitro prior to implantation, in terms of their ability to support
the cells within over prolonged periods of time and to exhibit and maintain their overall secretory properties Long-term cultures can be performed in perfusion bioreactors under
conditions simulating aspects of the in vivo environment
In certain studies, the bioreactors and support perfusion circuits were made compatible with a nuclear magnetic resonance spectrometer This allowed measuring intracel-lular metabolites, such as nucleotide triphosphates, as a function of culture conditions and time, without the need
I I I C O N S T R U C T T E C H N O L O G Y • 625
Trang 8to extract the encapsulated cells (Papas et al., 1999a, 1999b)
Such studies produce a comprehensive understanding of
the intrinsic tissue function in a well defi ned and controlled
environment prior to introducing the additional complexity
of host–implant interactions in in vivo experiments.
The secretory properties of tissue constructs can be
evaluated with low time resolution in simple static culture
experiments by changing the concentration of glucose in
the medium and measuring the secreted insulin In general,
a square wave of insulin concentration is implemented,
from basal to inducing basal conditions for insulin
secre-tion To evaluate the secretory response with a higher time
resolution, perfusion experiments need to be performed,
in which medium is fl owed around the tissue and secreted
insulin is assayed in the effl uent Again, a square wave
of glucose concentration is generally implemented By
com-paring the secretory dynamics of free and encapsulated
cells, one can ensure that the encapsulation material
does not introduce excessive time lags that might
compro-mise the secretory properties of the construct Indeed,
properly designed hydrogel microcapsules introduce only
minimal secretory time lags (Tziampazis and Sambanis,
1995; Sambanis et al., 2002).
Manufacturing Considerations
Fabrication of pancreatic substitutes of consistent
quality requires the use of cells that are also of consistent
quality Although with clonal, expandable cells this is a
rather straightforward issue, with islets isolated from human
and animal tissues there can be signifi cant variability in the
quantity and quality of the cells in the preparations With
islets from cadaveric human donors, the quality of the
iso-lates is assessed by microscopic observation, viability
stain-ing, and possibly a static insulin secretion test It is generally
recognized, however, that a quantitative, objective
assess-ment of islet quality would help improve the consistency of
the preparations and thus, possibly, the transplantation
outcome
It is conceivable that encapsulated cell systems could
be fabricated at a central location from which they are
dis-tributed to clinical facilities for implantation In this scheme,
preservation of the constructs for long-term storage,
inven-tory management, and, importantly, sterility control would
be essential Cryopreservation appears to be a promising
method for maintaining fabricated constructs for prolonged
time periods Although there have been signifi cant studies
on the cryopreservation of single cells and some tissues, the
problems pertaining to cryopreserving artifi cial tissues are
only beginning to be addressed Cryopreservation of
mac-roencapsulated systems is expected to be particularly
chal-lenging and has not been reported in the literature However,
βTC-cells encapsulated in alginate beads have been
pre-served successfully (Mukherjee et al., 2005; Song et al., 2005)
An especially promising approach involves using high
concentrations of cryoprotective agents so that water is
converted to a glassy, or vitrifi ed, state at low temperatures;
the absence of ice crystals in both the intracellular and extracellular domains appears helpful in maintaining not only cellular viability but also the structure and function of
the surrounding matrix (Mukherjee et al., 2005; Song et al.,
2005)
IV IN VIVO IMPLANTATION
This section highlights results from in vivo experiments
using the different confi gurations outlined earlier Results with encapsulated cell systems are presented fi rst Since
in vivo experiments with non-β-cells engineered for insulin
secretion are at present based mostly on in vivo gene therapy
approaches, these are described next Technologies for the
in vivo monitoring of cells and constructs and the issue of
implant retrievability are then discussed
Encapsulated Cell Systems
In vivo experiments with pancreatic substitutes are
numerous in small animals, limited in large animals, and few in humans Allogeneic and xenogeneic islets in hydrogel microcapsules implanted in diabetic mice and rats have generally restored normoglycemia for prolonged periods of
time In the early study of O’Shea et al (1984), islet allografts
encapsulated in APA membranes were implanted toneally in streptozotocin-induced diabetic rats Of the fi ve animals that received transplants, three remained normo-glycemic for more than 100 days One of these three animals remained normoglycemic 368 days postimplantation In the
intraperi-later study of Lum et al (1992), rat islets encapsulated in APA
membranes and implanted in streptozotocin diabetic mice restored normoglycemia for up to 308 days, with a mean xenograft survival time of 220 days With all recipients, nor-moglycemia was restored within two days postimplanta-tion Control animals receiving single injections of unencapsulated islets remained normoglycemic for less
than two weeks (O’Shea et al., 1984; Lum et al., 1992) More
recently, APA-encapsulated βTC6-F7 insulinomas restored
normoglycemia in diabetic rats for up to 60 days (Mamujee
et al., 1997), and APA-encapsulated βTC-tet insulinomas in
NOD mice for at least eight weeks (Black et al., 2006) In the
latter study, it was also observed that no host cell adherence occurred to microcapsules, and there were no signifi cant immune responses to the implant, with cytokine levels being similar to those of sham-operated controls These results are thus indicative of the potential use of an immu-noisolated continuous β-cell line for the treatment of diabe-
tes With the recently developed human cell line (Narushima
et al., 2005), experiments were performed with
unencapsu-lated cells transplanted into streptozotocin-induced betic severe combined immunodefi ciency mice Control of blood glucose levels started within two weeks postimplanta-tion, and mice remained normoglycemic for longer than 30
dia-weeks (Narushima et al., 2005) Besides rodents, long-term
restoration of normoglycemia with microencapsulated islets
Trang 9has been demonstrated in large animals, including
sponta-neously diabetic dogs, where normoglycemia was achieved
with canine islet allografts for up to 172 days (Soon-Shiong
et al., 1992), and monkeys, where in one animal porcine islet
xenografts normalized hyperglycemia for more than 150
days (Sun et al., 1992) More recently, one of the companies
working on islet encapsulation technology announced that
primate subjects in ongoing studies have continued to
exhibit improved glycemic regulation over a six-month
period after receiving microencapsulated porcine islet
transplants (MicroIslet Inc Press Release, August 7, 2006)
In vivo results with vascular devices are reportedly
mixed Implantation of devices containing allogeneic islets
as arteriovenous shunts in pancreatectomized dogs resulted
in 20–50% of the dogs becoming normoglycemic up to 10
weeks postimplantation without exogenous insulin
admin-istration When xenogeneic bovine or porcine islets were
used, only 10% of the dogs remained normoglycemic 10
weeks postimplantation All dogs were reported diabetic or
dead after 15 weeks (Sullivan et al., 1991) Recently, a
hollow-fi ber device composed of polyethylene-vinyl alcohol hollow-fi bers
and a poly-amino-urethane-coated, nonwoven
polytetra-fl uoroethylene fabric seeded with porcine islets provided
normalization of the blood glucose levels in totally
pancre-atectomized pigs when connected to the vasculature of the
animals (Ikeda et al., 2006) It should be noted, however, that
the overall interest in vascular devices has faded, due to the
surgical and blood coagulation challenges they pose
Although several hypotheses exist, the precise cause of
the eventual in vivo failure of encapsulated cell systems
remains unclear Encapsulation does not completely prevent
the immune recognition of the implant Although direct
cel-lular recognition is prevented, antigens shed by the cells as
a result of secretion and, more importantly, lysis in the
cap-sules eventually pass through the permselective barrier and
are recognized by the antigen-presenting cells of the host
For example, in one study, antibodies against islets in a
tubular diffusion chamber were detected in plasma two to
six weeks postimplantation, suggesting that islet antigens
crossed the membrane and stimulated antibody formation
in the host (Lanza et al., 1994) In another study,
alginate-encapsulated islets were lysed in vitro by nitric oxide
pro-duced by activated macrophages (Wiegand et al., 1993)
Passage of low-molecular-weight molecules cannot be
prevented by immunoprotective membranes imposing a
molecular weight cutoff on the order of 50 kDa It should be
noted that in one human study involving encapsulated
allo-geneic islets, the patient had to be provided with low levels
of immunosuppression (Soon-Shiong et al., 1994) In a more
recent report, also with encapsulated allografts implanted
peritoneally, type 1 diabetic patients remained
nonimmu-nosuppressed but were unable to withdraw exogenous
insulin (Calafi ore et al., 2006).
Nonspecifi c infl ammation may also occur around the
implant and develop into a fi brous capsule, reducing the
oxygen available to the cells within The fi brotic layer has been found to consist of several layers of fi broblasts and collagen with polymorphonuclear leukocytes, macrophages, and lymphocytes The surface roughness of the membrane may also trigger infl ammatory responses In one study, membranes with smooth outside surfaces exhibited a minimal fi brotic reaction 10 weeks postimplantation, regardless of the type of encapsulated cells, whereas rough surfaces elicited a fi brotic response even one week postim-
plantation (Lanza et al., 1991) Use of high-purity materials
also helps to minimize infl ammatory reactions If a material
is intrinsically infl ammatory, such as poly-l-lysine, it can be coated with a layer of noninfl ammatory material, such as alginate, to minimize the host’s reaction Such coverage may not be suffi ciently permanent, though, resulting in the eventual fi brosis of the implant Indeed, several investi-gators report improved results with plain alginate beads without a poly-l-lysine layer, especially when allogeneic cells are used in the capsules
Provision of nutrients to and removal of metabolites from encapsulated cells can be especially challenging
in vivo Normal pancreatic islets are highly vascularized
and thus well oxygenated There exists evidence that unencapsulated islets injected in the portal system of the liver become revascularized, which enhances their engraft-ment and function On the other hand, encapsulation prevents revascularization, so the implanted tissue is nour-ished by diffusion alone Promotion of vascularization around the immunoprotective membrane increases the
oxygenation of the implanted islets (Prokop et al., 2001)
Interestingly, transformed cells, such as the βTC3 line of mouse insulinomas, are more tolerant of hypoxic conditions than intact islets; such cells may thus function better
than islets in implanted capsules (Papas et al., 1996)
However, with transformed cells, too, enhanced ation increases the density of functional cells that can be effectively maintained within the implant volume Vascular-ization is dependent on the microarchitecture of the mate-rial, which should have pores 0.8–8.0 µm in size, allowing
oxygen-permeation of host endothelial cells (Brauker et al., 1992,
1995) Vascularization is also enhanced by the delivery of angiogenic agents, such as FGF-2 and VEGF, possibly with
controlled-release devices (Sakurai et al., 2003) Although
vascularization can be promoted around a cell-seeded device, improved success has been reported if a cell-free device is fi rst implanted and vascularized and the cells are then introduced One example of this procedure involved placing a cylindrical stainless steel mesh in the subcutane-ous space of rats, with the islets introduced 40 days later
(Pileggi et al., 2006) Replacement of a vascularized implant
is challenging, however, due to the bleeding that occurs A solution to this problem may entail the design of a device that can be emptied and refi lled with a suspension of cells
in an extracellular matrix without disturbing the housing and the associated vascular network
I V I N V I V O I M P L A N T A T I O N • 627
Trang 10Gene and Cell-Based Therapies
In vivo effi cacy studies with gene therapy and non-
β-cells genetically engineered for insulin secretion are
gener-ally limited to small animals Intraportal injection of
recombinant adenovirus expressing furin-compatible
insulin under the control of a glucose-responsive promoter
containing elements of the rat liver pyruvate kinase gene
restored near-normal glycemia in streptozotocin diabetic
rats for periods of 1–12 weeks (Thule and Liu, 2000) With
hepatic delivery of a recombinant adeno-associated virus
expressing a single-chain insulin analog under the control
of an l-type pyruvate kinase promoter, Lee and coworkers
(2000) controlled blood glucose levels in streptozotocin
dia-betic rats and NOD mice for periods longer than 20 weeks
However, transiently low blood glucose levels observed
three to fi ve hours after glucose loading indicated a
draw-back of the transcriptional regulation of insulin expression,
which may result in hypoglycemic episodes (Lee et al., 2000)
Possible approaches toward ameliorating this problem
include optimizing the number of glucose-regulatory and
insulin-sensing elements in the promoter (Jun and Yoon,
2005) and destabilizing the preproinsulin mRNA; the latter
has been shown to expedite signifi cantly the
down-regula-tion of secredown-regula-tion dynamics from transcripdown-regula-tionally controlled
cells on removal of the secretory stimulus (Tang and
Sam-banis, 2003)
In vivo gene therapy with small animals has also shown
success when the target cells for insulin expression were
intestinal endocrine K- or gastric G-cells Using a transgene
expressing human insulin under the control of the
glucose-dependent insulinotropic peptide (GIP) promoter, Cheung
et al (2000) expressed insulin specifi cally in gut K-cells of
transgenic mice, which protected them from developing
diabetes following STZ-mediated destruction of the native
β-cells Similarly, use of a tissue-specifi c promoter to express
insulin in gastric G-cells of mice resulted in insulin release
into circulation in response to meal-associated stimuli,
sug-gesting that G-cell insulin expression is benefi cial in the
amelioration of diabetes (Lu et al., 2005) Translation of
these approaches to adult animals and, eventually, humans,
requires the development of effective methods of gene
delivery to intestinal endocrine or gastric cells in vivo or the
development of effective ex vivo gene therapy approaches.
In Vivo Monitoring
Monitoring of the number and function of
insulin-secreting cells in vivo would provide valuable information
directly on the implant and possibly offer early indications
of implant failure Additionally, in animal experiments, the
ability to monitor an implant noninvasively reduces the
number of animals that are needed in the experimental
design and helps establish a critical link between
implanta-tion and endpoint physiologic effects, the latter commonly
being blood glucose levels and animal weight
Imaging techniques can provide unique insight into the
structure/function relationship of a construct in vitro and
in vivo There are several imaging modalities that have been
applied to monitor tissue-engineered constructs, including computed tomography (CT), positron emission tomogra-phy (PET), optical techniques, and nuclear magnetic reso-nance (NMR) imaging and spectroscopy Among these, NMR offers the unique advantage of providing information
on both construct integrity and function, without the need
to modify the cells genetically (e.g., through the expression
of green fl uorescent protein, used in optical methods) or the introduction of radioactive labels (e.g., PET agents)
Furthermore, since magnetic fi elds penetrate uniformly throughout the sample, NMR is ideally suited to monitor constructs implanted at deep-seated locations Its disad-vantage is its low sensitivity Whereas optical and radionu-clide techniques can detect tracer quantities, NMR detects metabolites that are available in the millimolar or, in some cases, submillimolar range
The ability to monitor noninvasively in vivo a pancreatic substitute by NMR was reported recently (Stabler et al.,
2005) Agarose disc-shaped constructs containing mouse insulinoma βTC3-cells were implanted in the peritoneal cavity of mice Construct integrity was visualized by MR imaging and the metabolic activity of the cells within by water-suppressed 1H NMR spectroscopy (Fig 42.3) Control experiments established that the total choline (TCho) reso-nance at 3.2 ppm, which is attributed to three choline metabolites, correlated positively and linearly with the number of viable cells within the construct, measured with
an independent assay To obtain the TCho signal in vivo
without interference from the surrounding host tissue, such
as peritoneal fat, the central agarose disc containing the cells had to be surrounded by cell-free agarose buffer zones This ensured that the MR signal arose only from the implanted cells, even as the construct moved due to animal breathing
A second problem that had to be resolved was that glucose diffusing into the construct produced a resonance that inter-fered with the TCho resonance at 3.2 ppm For this, a unique glucose resonance at 3.85 ppm was used to correct for the interference at 3.2 ppm so that a corrected signal, uniquely attributed to TCho, could be obtained The latter correlated positively and linearly with the number of viable cells, mea-sured with an independent assay, on the constructs postex-plantation (Fig 42.3) Hence, with the appropriate implanted construct architecture and signal processing, the number of viable cells in an implant could be followed in the same
animal as a function of time (Stabler et al., 2005).
Labeling of cells with magnetic nanoparticles, which can be detected by magnetic resonance, and genetically engineering cells so that they express a fl uorescent or lumi-nescent marker that can be optically detected are other methods being pursued to track the location and possibly
function of implanted cells in vivo It is expected that
devel-opment of robust monitoring methodologies will be helpful
Trang 11not only in experimental development studies but also in
eventual clinical applications
Retrievability
The issue of construct retrievability needs to be
consid-ered for all pertinent applications Useful lifetimes of
con-structs are limited, so repeated implantation of cells will be
required It is as yet unclear whether retrieval of constructs
will be necessary at the end of their useful lives Long-term
studies on the safety challenges posed by accumulated
implants in the host need to be carried out to address this
question
V CONCLUDING REMARKS
Tight glycemic regulation in insulin-dependent
diabet-ics signifi cantly improves their overall health and reduces
the long-term complications of the disease A pancreatic
substitute holds signifi cant promise at accomplishing this
in a relatively noninvasive way However, to justify the
improved outcome, a substitute needs to be not only effi
ca-cious in terms of insulin secretion, but also
immunologi-cally acceptable A number of approaches are being pursued
to address this obstacle and additionally develop constructs that can be manufactured at a clinically relevant scale However, as the problems are being thoroughly investigated, their solutions become more challenging Encapsulation in permselective barriers improves the immune acceptance of allo- and xenografts, but it is doubtful that encapsulation will, by itself, ensure the long-term survival and function of the implant in nonimmunosuppressed hosts The develop-ment of specifi c, benign immune suppression protocols that work in concert with encapsulation appears necessary Reducing the immunogenicity of the implanted cells and modifying them so that they better withstand the encapsu-
lation and in vivo environment are appropriate strategies
To ensure that substitutes can be fabricated at the necessary scale, methods to expand pancreatic islets in culture, to produce β-cells from stem cells, or to generate expandable β-cell lines with appropriate phenotypic characteristics need to be pursued In alternative approaches involving gene therapy of non-β-cells, or the ex vivo engineering of
non-β-cells retrieved surgically from the host, the major problem is not that of cell procurement or immune accep-tance but, rather, of ensuring precise regulation of insulin
cells/mL agarose implanted in the peritoneal cavity of a mouse (A) 1
H NMR image obtained with a surface coil The inner disc, containing the cells, is distinguishable from the surrounding cell-free buffer zone, implemented to exclude spectroscopic signal from the surrounding host
tissue (B) Localized, water-suppressed 1
H NMR spectrum from the cells contained in the inner disc Resonances due to total choline (TCho), glucose, and
lactate are clearly visible The time needed to collect the spectrum was 13 min (C) Correlation between the glucose-corrected TCho resonance at 3.2 ppm
and the viable cell number obtained postexplantation using the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide] assay Adapted from
Stabler et al (2005).
Trang 12secretion by glucose or other physiologic stimuli This poses
a different set of problems, which, however, are equally
challenging to those of β-cell procurement and immune
acceptance Methods for the preservation of substitutes and
for the noninvasive monitoring of their integrity and
func-tionality in vivo are integral parts of construct development
and characterization with regard to construct
manufactur-ing and assessment of in vivo effi cacy, respectively.
As in many aspects of life, with challenges come
oppor-tunities It is essential that multiple approaches be pursued
in parallel, because it is currently unclear which ones will
eventually develop into viable therapeutic procedures If more than one method evolves into a clinical application, this would be welcome news, because it may allow fl exibility
in the personalization of therapy For instance, in an adult type 2 insulin-dependent diabetic, use of an encapsulated allograft with low-level immunosuppression might consti-tute an appropriate therapeutic modality In a juvenile type
1 diabetic with aggressive autoimmunity, however, use of autologous genetically engineered non-β-cells, which are not recognized by the resident autoimmunity, may consti-tute the therapeutic method of choice
VI ACKNOWLEDGMENTS
The studies in the author’s and coinvestigators’ laboratories
referenced in this chapter were supported by grants from
the Georgia Tech/Emory Center for the Engineering of Living
Tissues (GTEC), a National Science Foundation Engineering
Research Center; and by grants from the National Institutes
of Health, EmTech Bio, and the Juvenile Diabetes Research Foundation international The author also wishes to thank Indra Neil Mukherjee and Heather Bara for critically review-ing the manuscript, as well as Drs Constantinidis and Thulé for helpful discussions
VII REFERENCES
Bigam, D L., and Shapiro, A J (2004) Pancreatic transplantation:
beta-cell replacement Curr Treat Options Gastroenterol 7, 329–341.
Black, S P., Constantinidis, I., Cui, H., Tucker-Burden, C., Weber, C J.,
and Safl ey, S A (2006) Immune responses to an encapsulated
alloge-neic islet beta-cell line in diabetic NOD mice Biochem Biophys Res
Commun 340, 236–243.
Bloch, K., Assa, S., Lazard, D., Abramov, N., Shalitin, S., Weintrob, N.,
Josefsberg, Z., Rapoport, M., and Vardi, P (1999) Neonatal pig islets
induce a lower T-cell response than adult pig islets in IDDM patients
Transplantation 67, 748–752.
Bloch, K., Papismedov, E., Yavriyants, K., Vorobeychik, M., Beer, S., and
Vardi, P (2006) Photosynthetic oxygen generator for bioartifi cial
pan-creas Tissue Eng 12, 337–344.
Blyszczuk, P., Czyz, J., Kania, G., Wagner, M., Roll, U., St-Onge, L., and
Wobus, A M (2003) Expression of Pax4 in embryonic stem cells
promotes differentiation of nestin-positive progenitor and
insulin-producing cells Proc Natl Acad Sci U.S.A 100, 998–1003.
Bonner-Weir, S., and Sharma, A (2002) Pancreatic stem cells
J Pathol 197, 519–526.
Bonner-Weir, S., Taneja, M., Weir, G C., Tatarkiewicz, K., Song, K H.,
Sharma, A., and O’Neil, J J (2000) In vitro cultivation of human islets
from expanded ductal tissue Proc Natl Acad Sci U.S.A 97,
7999–8004.
Brauker, J., Martinson, L A., Hill, R S., Young, S K., Carr-Brendel, V E.,
and Johnson, R C (1992) Neovascularization of immunoisolation
membranes: the effect of membrane architecture and encapsulated
tissue Transplant Proc 24, 2924.
Brauker, J H., Carr-Brendel, V E., Martinson, L A., Crudele, J.,
Johnston, W D., and Johnson, R C (1995) Neovascularization of
syn-thetic membranes directed by membrane microarchitecture
J Biomed Mater Res 29, 1517–1524.
Calafi ore, R., Basta, G., Luca, G., Lemmi, A., Montanucci, M P.,
Calabrese, G., Racanicchi, L., Mancuso, F., and Brunetti, P (2006)
Microencapsulated pancreatic islet allografts into
nonimmunosup-pressed patients with type 1 diabetes: fi rst two cases Diabetes Care 29,
Chen, G., Hohmeier, H E., Gasa, R., Tran, V V., and Newgard, C B
(2000) Selection of insulinoma cell lines with resistance to
interleukin-1beta- and gamma-interferon-induced cytotoxicity Diabetes 49,
562–570.
Cheng, S Y., Gross, J., and Sambanis, A (2004) Hybrid pancreatic tissue substitute consisting of recombinant insulin-secreting cells and
glucose-responsive material Biotechnol Bioeng 87, 863–873.
Cheng, S Y., Constantinidis, I., and Sambanis, A (2006) Use of responsive material to regulate insulin release from constitutively
glucose-secreting cells Biotechnol Bioeng 93, 1079–1088.
Cheung, A T., Dayanandan, B., Lewis, J T., Korbutt, G S., Rajotte, R V., Bryer-Ash, M., Boylan, M O., Wolfe, M M., and Kieffer, T J (2000)
Glucose-dependent insulin release from genetically engineered K cells
Science 290, 1959–1962.
Clark, S A., Quaade, C., Constandy, H., Hansen, P., Halban, P., Ferber, S., Newgard, C B., and Normington, K (1997) Novel insulinoma cell lines produced by iterative engineering of GLUT2, glucokinase, and
human insulin expression Diabetes 46, 958–967.
Creutzfeldt, W (1974) [Clinical signifi cance of gastrointestinal
hor-mones] Verh Dtsch Ges Inn Med 80, 330–338.
Cruise, G M., Hegre, O D., Lamberti, F V., Hager, S R., Hill, R., Scharp,
D S., and Hubbell, J A (1999) In vitro and in vivo performance
of porcine islets encapsulated in interfacially photopolymerized
poly(ethylene glycol) diacrylate membranes Cell Transplant 8,
293–306.
Delaunay, C., Darquy, S., Honiger, J., Capron, F., Rouault, C., and Reach,
G (1998) Glucose-insulin kinetics of a bioartifi cial pancreas made of
an AN69 hydrogel hollow fi ber containing porcine islets and implanted
in diabetic mice Artif Organs 22, 291–299.
Drucker, D J (2002) Biological actions and therapeutic potential of the
glucagon-like peptides Gastroenterology 122, 531–544.
Trang 13Efrat, S (1998) Cell-based therapy for insulin-dependent diabetes
mel-litus Eur J Endocrinol 138, 129–133.
Efrat, S., Linde, S., Kofod, H., Spector, D., Delannoy, M., Grant, S.,
Hanahan, D., and Baekkeskov, S (1988) Beta-cell lines derived from
transgenic mice expressing a hybrid insulin gene-oncogene Proc Natl
Acad Sci U.S.A 85, 9037–9041.
Efrat, S., Leiser, M., Surana, M., Tal, M., Fusco-Demane, D., and Fleischer,
N (1993) Murine insulinoma cell line with normal glucose-regulated
insulin secretion Diabetes 42, 901–907.
Efrat, S., Fusco-DeMane, D., Lemberg, H., al Emran, O., and Wang, X
(1995) Conditional transformation of a pancreatic beta-cell line derived
from transgenic mice expressing a tetracycline-regulated oncogene
Proc Natl Acad Sci U.S.A 92, 3576–3580.
Finegood, D T., Scaglia, L., and Bonner-Weir, S (1995) Dynamics of
beta-cell mass in the growing rat pancreas Estimation with a simple
mathematical model Diabetes 44, 249–256.
Gross, J D., Constantinidis, I., and Sambanis, A (2007) Modeling of
encapsulated cell systems J Theor Biol 244, 500–510.
Heald, K A., Carless N., Jay, T R., Boucher, N., and Downing, R (1999)
Expression of the GALalpha(1–3)GAL epitope on pig islets J Mol Med
77, 169–171.
Hill, R S., Cruise, G M., Hager, S R., Lamberti, F V., Yu, X., Garufi s,
C L., Yu, Y., Mundwiler, K E., Cole, J F., Hubbell, J A., Hegre, O D.,
and Scharp, D W (1997) Immunoisolation of adult porcine islets for
the treatment of diabetes mellitus The use of photopolymerizable
polyethylene glycol in the conformal coating of mass-isolated porcine
islets Ann N.Y Acad Sci 831, 332–343.
Hughes, S D., Johnson, J H., Quaade, C., and Newgard, C B (1992)
Engineering of glucose-stimulated insulin secretion and biosynthesis
in non-islet cells Proc Natl Acad Sci U.S.A 89, 688–692.
Hughes, S D., Quaade, C., Johnson, J H., Ferber, S., and Newgard, C B
(1993) Transfection of AtT-20ins cells with GLUT-2 but not GLUT-1
confers glucose-stimulated insulin secretion Relationship to glucose
metabolism J Biol Chem 268, 15205–15212.
Ikeda, H., Kobayashi, N., Tanaka, Y., Nakaji, S., Yong, C., Okitsu, T.,
Oshita, M., Matsumoto, S., Noguchi, H., Narushima, M., Tanaka, K.,
Miki, A., Rivas-Carrillo, J D., Soto-Gutierrez, A., Navarro-Alvarez, N.,
Tanaka, K., Jun, H S., Tanaka, N., and Yoon, J W (2006) A newly
developed bioartifi cial pancreas successfully controls blood glucose
in totally pancreatectomized diabetic pigs Tissue Eng 12, 1799–1809.
Jun, H S., and Yoon, J W (2005) Approaches for the cure of type 1
dia-betes by cellular and gene therapy Curr Gene Ther 5, 249–262.
Kieffer, T J., and Habener, J F (1999) The glucagon-like peptides
Endocr Rev 20, 876–913.
Knaack, D., Fiore, D M., Surana, M., Leiser, M., Laurance, M.,
Fusco-DeMane, D., Hegre, O D., Fleischer, N., and Efrat, S (1994) Clonal
insulinoma cell line that stably maintains correct glucose
responsive-ness Diabetes 43, 1413–1417.
Kojima, H., Fujimiya, M., Matsumura, K., Younan, P., Imaeda, H., Maeda,
M., and Chan, L (2003) NeuroD-betacellulin gene therapy induces islet
neogenesis in the liver and reverses diabetes in mice Nat Med 9,
596–603.
Lannoy, V J., Decaux, J F., Pierreux, C E., Lemaigre, F P., and Rousseau,
G G (2002) Liver glucokinase gene expression is controlled by the
onecut transcription factor hepatocyte nuclear factor-6 Diabetologia
45, 1136–1141.
Lanza, R P., Butler, D H., Borland, K M., Staruk, J E., Faustman, D L.,
Solomon, B A., Muller, T E., Rupp, R G., Maki, T., Monaco, A P., et al
(1991) Xenotransplantation of canine, bovine, and porcine islets in
diabetic rats without immunosuppression Proc Natl Acad Sci U.S.A
88, 11100–11104.
Lanza, R P., Sullivan, S J., and Chick, W L (1992) Perspectives in
dia-betes Islet transplantation with immunoisolation Diabetes 41,
1503–1510.
Lanza, R P., Beyer, A M., and Chick, W L (1994) Xenogenic humoral responses to islets transplanted in biohybrid diffusion chambers
Transplantation 57, 1371–1375.
Lechner, A., Nolan, A L., Blacken, R A., and Habener, J F (2005)
Redif-ferentiation of insulin-secreting cells after in vitro expansion of adult
human pancreatic islet tissue Biochem Biophys Res Commun 327,
581–588.
Lee, H C., Kim, S J., Kim, K S., Shin, H C., and Yoon, J W (2000) Remission in models of type 1 diabetes by gene therapy using a single-
chain insulin analogue Nature 408, 483–488.
Lu, Y.-C., Sternini, C., Rozengurt, E., and Zhokova, E (2005) Release of transgenic human insulin from gastric G cells: a novel approach for the
amelioration of diabetes Endocrinology 146, 2610–2619.
Lum, Z P., Krestow, M., Tai, I T., Vacek, I., and Sun, A M (1992) grafts of rat islets into diabetic mice An evaluation of new smaller
Xeno-capsules Transplantation 53, 1180–1183.
Lumelsky, N., Blondel, O., Laeng, P., Velasco, I., Ravin, R., and McKay, R (2001) Differentiation of embryonic stem cells to insulin-secreting
structures similar to pancreatic islets Science 292, 1389–1394.
Lundberg, P., and Kuchel, P W (1997) Diffusion of solutes in agarose and alginate gels: 1H and 23Na PFGSE and 23Na TQF NMR
studies Magn Reson Med 37, 44–52.
Mamujee, S N., Zhou, D., Wheeler, M B., Vacek, I., and Sun, A M (1997) Evaluation of immunoisolated insulin-secreting beta TC6-F7
cells as a bioartifi cial pancreas Ann Transplant 2, 27–32.
May, M H., and Sefton, M V (1999) Conformal coating of small
parti-cles and cell aggregates at a liquid–liquid interface Ann N.Y Acad Sci
875, 126–134.
Mikos, A., Papadaki, M., Kouvroukoglou, S., Ishaug, S., and Thomson,
R (1994) Mini-review: islet transplantation to create a bioartifi cial
pan-creas Biotechnol Bioeng 43, 673–677.
Miyazaki, J., Araki, K., Yamato, E., Ikegami, H., Asano, T., Shibasaki, Y., Oka, Y., and Yamamura, K (1990) Establishment of a pancreatic beta cell line that retains glucose-inducible insulin secretion: special refer-
ence to expression of glucose transporter isoforms Endocrinology 127,
Nerem, R M., and Sambanis, A (1995) Tissue engineering: from biology
to biological substitutes Tissue Eng 1, 3–13.
V I I R E F E R E N C E S • 631
Trang 14O’Shea, G M., Goosen, M F., and Sun, A M (1984) Prolonged survival
of transplanted islets of Langerhans encapsulated in a biocompatible
membrane Biochim Biophys Acta 804, 133–136.
Olson, D E., Paveglio, S A., Huey, P U., Porter, M H., and Thule, P M
(2003) Glucose-responsive hepatic insulin gene therapy of
spontane-ously diabetic BB/Wor rats Hum Gene Ther 14, 1401–1413.
Ouziel-Yahalom, L., Zalzman, M., Anker-Kitai, L., Knoller, S., Bar, Y.,
Glandt, M., Herold, K., and Efrat, S (2006) Expansion and
redifferentia-tion of adult human pancreatic islet cells Biochem Biophys Res
Commun 341, 291–298.
Papas, K K., Long, R C Jr., Constantinidis, I., and Sambanis, A (1996)
Effects of oxygen on metabolic and secretory activities of beta TC3 cells
Biochim Biophys Acta 1291, 163–166.
Papas, K K., Long, R C., Jr., Sambanis, A., and Constantinidis, I (1999a)
Development of a bioartifi cial pancreas: I Long-term propagation and
basal and induced secretion from entrapped betaTC3 cell cultures
Bio-technol Bioeng 66, 219–230.
Papas, K K., Long, R C., Jr., Sambanis, A., and Constantinidis, I (1999b)
Development of a bioartifi cial pancreas: II Effects of oxygen on
long-term entrapped betaTC3 cell cultures Biotechnol Bioeng 66,
231–237.
Pileggi, A., Molano, R D., Ricordi, C., Zahr, E., Collins, J., Valdes, R., and
Inverardi, L (2006) Reversal of diabetes by pancreatic islet
transplanta-tion into a subcutaneous, neovascularized device Transplantatransplanta-tion 81,
1318–1324.
Prevost, P., Flori, S., Collier, C., Muscat, E., and Rolland, E (1997)
Appli-cation of AN69 hydrogel to islet encapsulation Evaluation in
strepto-zotocin-induced diabetic rat model Ann N.Y Acad Sci 831, 344–349.
Prokop, A (2001) Bioartifi cial pancreas: materials, devices, function,
and limitations Diabetes Technol Ther 3, 431–449.
Prokop, A., Kozlov, E., Nun Non, S., Dikov, M M., Sephel, G C., Whitsitt,
J S., and Davidson, J M (2001) Towards retrievable vascularized
bio-artifi cial pancreas: induction and long-lasting stability of polymeric
mesh implant vascularized with the help of acidic and basic fi broblast
growth factors and hydrogel coating Diabetes Technol Ther 3,
245–261.
Rajagopal, J., Anderson, W J., Kume, S., Martinez, O I., and Melton,
D A (2003) Insulin staining of ES cell progeny from insulin uptake
Science 299, 363.
Ramiya, V K., Maraist, M., Arfors, K E., Schatz, D A., Peck, A B., and
Cornelius, J G (2000) Reversal of insulin-dependent diabetes using
islets generated in vitro from pancreatic stem cells Nat Med 6,
278–282.
Rayat, G R., Rajotte, R V., Elliott, J F., and Korbutt, G S (1998)
Expres-sion of Gal alpha(1,3)gal on neonatal porcine islet beta-cells and
sus-ceptibility to human antibody/complement lysis Diabetes 47, 1406–
1411.
Sakurai, T., Satake, A., Nagata, N., Gu, Y., Hiura, A., Doo-Hoon, K., Hori,
H., Tabata, Y., Sumi, S., and Inoue, K (2003) The development of new
immunoisolatory devices possessing the ability to induce
neovascular-ization Cell Transplant 12, 527–535.
Sambanis, A (2000) Engineering challenges in the development of an
encapsulated cell system for treatment of type 1 diabetes Diabetes
Technol Therap 2, 81–89.
Sambanis, A., and Tan, S A (1999) Quantitative modeling of limitations
caused by diffusion In “Methods in Molecular Medicine, Vol 18: Tissue
Engineering Methods and Protocols” (J R Morgan and M L Yarmush,
eds.) Humana Press, Totowa, NJ.
Sambanis, A., Stephanopoulos, G., Sinskey, A J., and Lodish, H F
(1990) Use of regulated secretion in protein production from animal
cells: an evaluation with the AtT-20 model cell line Biotechnol Bioeng
35, 771–780.
Sambanis, A., Tang, S.-C., Cheng, S.-Y., Stabler, C L., Long, R C J., and Constantinidis, I (2002) Core technologies in tissue engineering and
their application to the bioartifi cial pancreas In “Tissue Engineering
for Therapeutic Use” (Y Ikada, Y Umakoshi and T Hotta, eds.),
pp 5–18 Elsevier, Boston.
Schirra, J., Katschinski, M., Weidmann, C., Schafer, T., Wank, U., Arnold, R., and Goke, B (1996) Gastric emptying and release of incretin hor-
mones after glucose ingestion in humans J Clin Invest 97, 92–103.
Sefton, M., and Kharlip, L (1994) Insulin release from rat pancreatic
islets microencapsulated in a HEMA-MMA polyacrylate In “Pancreatic
Islet Transplantation Volume III: Immunoisolation of Pancreatic Islets”
(R Lanza and W Chick, eds.) RG Landes, Georgetown, TX.
Sefton, M V., May, M H., Lahooti, S., and Babensee, J E (2000) Making microencapsulation work: conformal coating, immobilization gels and
in vivo performance J Controlled Release 65, 173–186.
Shapiro, A M., Ryan, E A., and Lakey, J R (2001a) Clinical islet
trans-plant — state of the art Transtrans-plant Proc 33, 3502–3503.
Shapiro, A M., Ryan, E A., and Lakey, J R (2001b) Diabetes Islet cell
transplantation Lancet 358 (Suppl), S21.
Shapiro, A M., Ryan, E A., and Lakey, J R (2001c) Pancreatic islet
transplantation in the treatment of diabetes mellitus Best Pract Res
Clin Endocrinol Metab 15, 241–264.
Simpson, N E., Khokhlova, N., Oca-Cossio, J A., McFarlane, S S., Simpson, C P., and Constantinidis, I (2005) Effects of growth regula- tion on conditionally transformed alginate-entrapped insulin secreting
cell lines in vitro Biomaterials 26, 4633–4641.
Song, Y C., Chen, Z Z., Mukherjee, N., Lightfoot, F G., Taylor, M J., Brockbank, K G., and Sambanis, A (2005) Vitrifi cation of tissue-
engineered pancreatic substitute Transplant Proc 37, 253–255.
Soon-Shiong, P., Feldman, E., Nelson, R., Heintz, R., Merideth, N., Sandford, P., Zheng, T., and Komtebedde, J (1992) Long-term reversal
of diabetes in the large animal model by encapsulated islet
transplanta-tion Transplant Proc 24, 2946–2947.
Soon-Shiong, P., Heintz, R E., Merideth, N., Yao, Q X., Yao, Z., Zheng,
T., Murphy, M., Moloney, M K., Schmehl, M., Harris, M., et al (1994)
Insulin independence in a type 1 diabetic patient after encapsulated
islet transplantation Lancet 343, 950–951.
Soria, B., Roche, E., Berna, G., Leon-Quinto, T., Reig, J A., and Martin,
F (2000) Insulin-secreting cells derived from embryonic stem cells
nor-malize glycemia in streptozotocin-induced diabetic mice Diabetes 49,
157–162.
Stabler, C., Wilks, K., Sambanis, A., and Constantinidis, I (2001) The
effects of alginate composition on encapsulated betaTC3 cells
Bio-materials 22, 1301–1310.
Stabler, C L., Long, R C., Jr., Constantinidis, I., and Sambanis, A (2005)
In vivo noninvasive monitoring of a tissue-engineered construct using
1H-NMR spectroscopy Cell Transplant 14, 139–149.
Sullivan, S J., Maki, T., Borland, K M., Mahoney, M D., Solomon, B A., Muller, T E., Monaco, A P., and Chick, W L (1991) Biohybrid artifi cial pancreas: long-term implantation studies in diabetic, pancreatecto-
mized dogs Science 252, 718–721.
Sun, A M., Vacek, I., Sun, Y L., Ma, X., and Zhou, D (1992) In vitro and
in vivo evaluation of microencapsulated porcine islets Asaio J 38,
125–127.
Trang 15Tang, S.-C., and Sambanis, A (2003a) Preproinsulin mRNA engineering
and its application to the regulation of insulin secretion from human
hepatomas FEBS Lett 537, 193–197.
Tang, S C., and Sambanis, A (2003b) Development of genetically
engi-neered human intestinal cells for regulated insulin secretion using
rAAV-mediated gene transfer Biochem Biophys Res Commun 303,
645–652.
Tang, S C., and Sambanis, A (2004) Differential rAAV2 transduction
effi ciencies and insulin secretion profi les in pure and coculture models
of human enteroendocrine L-cells and enterocytes J Gene Med 6,
1003–1013.
Thule, P M., and Liu, J M (2000) Regulated hepatic insulin gene
therapy of STZ-diabetic rats Gene Ther 7, 1744–1752.
Thule, P M., Liu, J., and Phillips, L S (2000) Glucose-regulated
produc-tion of human insulin in rat hepatocytes Gene Ther 7, 205–214.
Thule, P M., Campbell, A G., Kleinhenz, D J., Olson, D E.,
Boutwell, J J., Sutliff, R L., and Hart, C M (2006) Hepatic insulin gene
therapy prevents deterioration of vascular function and improves
adi-pocytokine profi le in STZ-diabetic rats Am J Physiol Endocrinol
Metab 290, E114–E122.
Todorov, I., Omori, K., Pascual, M., Rawson, J., Nair, I., Valiente, L.,
Vuong, T., Matsuda, T., Orr, C., Ferreri, K., Smith, C V., Kandeel, F., and
Mullen, Y (2006) Generation of human islets through expansion and
differentiation of non-islet pancreatic cells discarded (pancreatic
discard) after islet isolation Pancreas 32, 130–138.
Tziampazis, E., and Sambanis, A (1995) Tissue engineering of a artifi cial pancreas: modeling the cell environment and device func -
bio-tion Biotechnol Prog 11, 115–126.
Wang, S., Liu, J., Li, L., and Wice, B M (2004) Individual subtypes of enteroendocrine cells in the mouse small intestine exhibit unique pat-
terns of inositol 1,4,5-trisphosphate receptor expression J Histochem
Cytochem 52, 53–63.
Wiegand, F., Kroncke, K D., and Kolb-Bachofen, V (1993) phage-generated nitric oxide as cytotoxic factor in destruction of alginate-encapsulated islets Protection by arginine analogs and/or
Macro-coencapsulated erythrocytes Transplantation 56, 1206–1212.
Wu, H., Avgoustiniatos, E S., Swette, L., Bonner-Weir, S., Weir, G C., and
Colton, C K (1999) In situ electrochemical oxygen generation with an
immunoisolation device Ann N.Y Acad Sci 875, 105–125.
Yanagita, M., Nakayama, K., and Takeuchi, T (1992) Processing of mutated proinsulin with tetrabasic cleavage sites to bioactive insulin in
the nonendocrine cell line, COS-7 FEBS Lett 311, 55–59.
Yanagita, M., Hoshino, H., Nakayama, K., and Takeuchi, T (1993) cessing of mutated proinsulin with tetrabasic cleavage sites to mature
Pro-insulin refl ects the expression of furin in nonendocrine cell lines
Endo-crinology 133, 639–644.
V I I R E F E R E N C E S • 633
Trang 16I Introduction
II Engineering to Generate Insulin-Producing
Cells
III Engineering to Improve Islet Survival
IV Vectors for Engineering Islets and Beta-Cells
V Conclusion
VI References
Engineering Pancreatic Beta-Cells
Hee-Sook Jun and Ji-Won Yoon
Principles of Tissue Engineering, 3 rd Edition
ed by Lanza, Langer, and Vacanti
Copyright © 2007, Elsevier, Inc.
All rights reserved.
I INTRODUCTION
The use of islet transplantation as a treatment for
dia-betes has been hampered by the limited availability of
human islets; therefore, new sources of insulin-producing
cells are needed Expansion of beta-cells by the generation
of reversibly immortalized beta-cells and creation of
insulin-producing cells by exogenous expression of insulin in
non-beta-cells have been investigated as new sources of
beta-cells Recently, embryonic and adult stem cells or
pan-creatic progenitor cells have been engineered to
differenti-ate into insulin-producing cells, demonstrating the possible
use of these cells for betacell replacement Despite signifi
-cant progress, further studies are needed to generate truly
functional insulin-producing cells In addition, the
engi-neering of beta-cells to protect them from immune attack
and to improve viability has been tried Although the
useful-ness of engineered beta-cells has yet to be clinically proven,
studies utilizing different engineering strategies and careful
analysis of the resulting insulin-producing cells may offer
potential methods to cure diabetes
Diabetes mellitus is a metabolic disease characterized
by uncontrolled hyperglycemia, which results in long-term
clinical problems, including retinopathy, neuropathy,
nephropathy, and heart disease Diabetes affects over 150
million people worldwide and is considered an epidemic of
the 21st century Blood glucose homeostasis is controlled by
endocrine beta-cells, located in the islets of Langerhans in
the pancreas When the concentration of blood glucose rises after a meal, insulin is produced and released from beta-cells Insulin then induces glucose uptake by cells in the body and converts glucose to glycogen in the liver When blood glucose concentration becomes low, glycogen is broken down to glucose in the liver and glucose is released into the blood
There are two major forms of diabetes: type 1 diabetes, also known as insulin-dependent diabetes mellitus, and type 2 diabetes, also known as non-insulin-dependent dia-betes mellitus Both types are thought to result from a reduc-tion in the number of insulin-producing beta-cells and defi cits in beta-cell function In type 1 diabetes, beta-cells are destroyed by autoimmune responses, resulting in a lack
of insulin (reviewed in Adorini et al., 2002; Yoon and Jun,
2005) In type 2 diabetes, both inadequate beta-cell function and insulin resistance of peripheral tissues contribute to the development of hyperglycemia, leading to eventual reduc-tion in the number of beta-cells (reviewed in LeRoith, 2002) Intensive exogenous insulin therapy has been used for the treatment of type 1 diabetes, but it does not restore the tight control of blood glucose levels or completely prevent the development of complications In addition, multiple daily injections are cumbersome and sometimes cause poten-tially life-threatening hypoglycemia Islet transplantation has been considered an alternative and safe method for the
treatment of diabetes (reviewed in Hatipoglu et al., 2005)
Trang 17636 C H A P T E R F O R T Y - T H R E E • E N G I N E E R I N G P A N C R E A T I C B E T A - C E L L S
With the improvement of islet isolation techniques, the
success rate for independence from exogenous insulin is
increasing However, the lack of suffi cient islets to meet the
demands of patients and the side effects of
immunosup-pressive drugs that are required to prevent alloimmune and
autoimmune attack against islet grafts are the major
limita-tions of islet transplantation Therefore, various alternative
sources of insulin-producing cells are being investigated to
provide a suffi cient supply for the treatment of type 1
diabetes
In this chapter, we discuss the use of cell engineering to
produce and expand insulin-producing beta-cells; to create
insulin-producing cells from non-beta-cells, embryonic
stem cells, and adult stem cells; and to improve islet graft
survival Due to the publisher’s restrictions, we are unable
to cite all the references for primary data
II ENGINEERING TO GENERATE
INSULIN-PRODUCING CELLS
Engineering Pancreatic Beta-Cells
The pancreas is composed of endocrine and exocrine
tissues The endocrine pancreas occupies less than 5% of
the pancreatic tissue mass and is composed of cell clusters
called the islets of Langerhans The islets of Langerhans
contain insulin-producing beta-cells (about 80% of cells in
the islets), glucagon-producing alpha-cells,
somatostatin-producing delta-cells, and pancreatic
polypeptide-produc-ing cells The exocrine pancreas occupies more than
95% of the pancreas and is composed of ascinar and
ductal cells, which produce digestive enzymes The beta-cell
mass is dynamic and increases in response to
environmen-tal changes such as pancreatic injury and physiological
changes such as insulin resistance In addition, mature
beta-cells can replicate throughout life, although at a low
level
One approach to produce beta-cells for replacement
therapy is to expand mature beta-cells in vitro However,
because mature beta-cells have limited proliferative
capac-ity in culture, the expression of oncogenes has been tried as
a method to establish beta-cell lines The expression of
simian virus (SV) 40 large T antigen in beta-cells under the
control of the tet-on and tet-off regulatory system in
trans-genic mice resulted in a stable beta-cell line that could be
expanded in vitro These cells produced less insulin in the
transformed state when T antigen was expressed, but insulin
production increased after growth was arrested by cessation
of T antigen expression, and insulin secretion was regulated
as in normal mouse islets When these cells were
trans-planted into streptozotocin-induced diabetic mice, the mice
became normoglycemic, and normoglycemia was
main-tained for a prolonged time, without any treatment to
prevent oncogene expression (Milo-Landesman et al., 2001)
In addition to beta-cell expansion, cell engineering has
been used to improve beta-cell function Rat insulinoma
cells that showed decreased glucose-responsive insulin secretion were transfected with a plasmid encoding a mutated form of GLP-1 that is resistant to the degrading enzyme dipeptidyl-peptidase IV These engineered cells had increased insulin secretion in response to glucose, as com-
pared with untransfected control cells (Islam et al., 2005).
Expansion of human primary pancreatic islet cells has also been tried Primary adult islet cells could be stimulated
to divide when grown on an extracellular matrix in the ence of hepatocyte growth factor/scatter factor, but growth was arrested after 10–15 cell divisions, due to cellular senes-
pres-cence (Beattie et al., 1999) Transformation of adult human
pancreatic islets with a retroviral vector expressing SV40 large T antigen and H-rasVal 12 oncogenes resulted in extended life span, but eventually the cells entered a crisis phase fol-lowed by altered morphology, lack of proliferation, and cell death, suggesting that immortalization of human beta-cells
is more diffi cult than that of rodent beta-cells However, introduction of human telomerase reverse transcriptase (hTERT) resulted in successful immortalization (Halvorsen
et al., 1999), because human cells do not express telomerase
This immortalized cell line, βlox5, initially expressed low levels of insulin, but insulin production subsequently fell to undetectable levels as a result of the loss of expression of key insulin gene transcription factors A combination of the introduction of a beta-cell transcription factor (Pdx-1), treatment with exendin-4 (a glucagon-like peptide-1 [GLP-1] homolog), and cell–cell contact was required to recover beta-cell differentiated function and glucose-responsive
insulin production (de la Tour et al., 2001) However, Pdx-1
expression in this cell line resulted in a signifi cant decrease
in the growth rate of the cells When streptozotocin-induced diabetic animals were transplanted with these Pdx-1-expressing cells, substantial levels of circulating human C-peptide were detected and diabetes was remitted However, 10% of the animals developed tumors, even though the oncogenes and hTERT gene had been fl oxed by loxP sites so that they could be deleted by expression of Cre recombi-nase This suggests that the Cre-expressing adenovirus and/
or Cre-mediated deletion of the oncogenes was ineffi cient
(de la Tour et al., 2001).
The limitations of previously engineered beta-cell lines point to a need for a human beta-cell line that is func-tionally equivalent to primary beta-cells, can be expanded indefi nitely, and can be rendered nontumorigenic In another approach to establish a reversibly immortalized human beta-cell line, human islets were transduced with a combination of retroviral vectors expressing SV40 T antigen, hTERT, and enhanced green fl uorescent protein to immor-talize and mark terminally differentiated pancreatic beta-cells These genes were fl oxed by loxP sites to allow excision
of the immortalizing genes Among 271 clones screened for tumorigenicity, 253 clones were selected for further study, and only one of these (NAKT-15) expressed insulin and the necessary beta-cell transcription factors, such as Isl-1, Pax-
Trang 186, Nkx6.1, Pdx-1, prohormone convertases, and secretory
granule proteins Addition of factors that enhance insulin
expression and secretion during culture of the beta-cell line,
such as troglitazone, a peroxisome proliferator-activated
receptor-γ activator, and nicotinamide, helped to maintain
the function of beta-cells, and culture of these cells on
Matrigel matrix facilitated aggregate formation Removal of
the immortalizing genes by Cre recombinase expression
stopped cell proliferation and increased the expression of
beta-cell-specifi c transcription factors, resulting in
rever-sion of the cells These reverted NAKT-15 cells were
func-tionally similar to normal human islets with respect to
insulin secretion in response to glucose and nonglucose
secretagogues, although the insulin content and amount of
secreted insulin were lower than for human islets However,
NAKT-15 cells were able to remit diabetes and clear
exoge-nous glucose when transplanted into diabetic severe
com-bined immunodefi ciency (SCID) mice The insulin content
of these cells was higher in vivo than in vitro, suggesting that
the microenvironment may enhance cellular differentiation
(Narushima et al., 2005).
For clinical application of reversibly immortalized
human beta-cells, safety issues, particularly tumorigenicity,
should be considered Reducing or eliminating
tumorige-nicity may be possible by using multiple selection
proce-dures In the case of NAKT-15 cells, nontumorigenic clones
were fi rst selected by screening for tumor formation in SCID
mice After infection of Cexpressing adenovirus to
re-move the SV40 T antigen and hTERT, SV40T-negative cells
were selected in the presence of a neomycin analog (the
neomycin-resistance gene was positioned to be expressed
after the loxP-fl anked genes were deleted), and
hTERT-neg-ative cells were selected by purifi cation of enhanced green
fl uorescent protein-negative cells Finally,
SV40T/hTERT-negative cells were selected by the addition of ganciclover,
because the cells had been transduced with a suicide gene,
herpes simplex thymidine kinase, which renders them
susceptible to ganciclover These multiple selection
pro-cedures resulted in no tumor development in SCID mice
when reverted NAKT-15 cells were transplanted (Narushima
et al., 2005), although the possibility of tumorigenesis could
not be completely eliminated Nevertheless, there are
advan-tages of reversibly immortalized human beta-cells as
com-pared with primary beta-cells They can be easily expanded
to obtain suffi cient cells for transplantation and genetically
manipulated in vitro prior to transplantation, for example,
to confer resistance to immune attack
Establishment of insulin-producing beta-cell lines by
reversible immortalization of primary islets is a promising
approach for replacing insulin injections, for a beta-cell line
can provide an abundant source of beta-cells for
transplan-tation In addition, beta-cell lines can be genetically
mani-pulated to improve their function and survival However, the
functionality of the cell lines and safety issues remain to be
further studied
Engineering Surrogate Beta-Cells
Non-beta-cells that are genetically engineered to produce insulin may have an advantage over intact islets or engineered beta-cells for transplantation therapy, because non-beta-cells should not be recognized by beta-cell-specifi c autoimmune responses Pancreatic beta-cells have unique characteristics specifi c to the production of insulin, such as specifi c peptidases, glucose-sensing systems, and secretory granules that can release insulin promptly by exocytosis in response to extracellular glucose levels There-fore, the ideal target cell to engineer for insulin production would be non-beta-cells possessing similar characteristics
A variety of cell types, including fi broblasts, hepatocytes, neuroendocrine cells, and muscle cells, have been engi-neered to produce insulin, with varying degrees of success
(reviewed in Xu et al., 2003; Yoon and Jun, 2002).
Neuroendocrine cells have received considerable tion because they have characteristics similar to those of beta-cells and contain components of the regulated secre-tory pathway, including prohormone convertases 2 and 3 and secretory granules A mouse corticotrophic cell line derived from the anterior pituitary, AtT20, expressed active insulin after transfection with the insulin gene under the control of a viral or metallothionein promoter but lacked glucose responsiveness Cotransfection of genes encoding glucose transporter (GLUT)2 and glucokinase conferred glucose-responsive insulin secretion in insulin-expressing AtT20 cells Transgenic expression of insulin in the interme-diate lobe of the pituitary of nonobese diabetic (NOD) mice under the control of the pro-opiomelanocortin promoter resulted in the production of biologically active insulin Transplantation of this insulin-producing pituitary tissue into diabetic NOD mice restored normoglycemia, but insulin secretion was not properly regulated by glucose Engineer-ing primary rat pituitary cells to coexpress GLP-1 receptor and human insulin resulted in GLP-1-induced insulin secre-
atten-tion (Wu et al., 2003).
Intestinal K-cells have been explored as possible gate beta-cells K-cells are endocrine cells located in the gut that secrete the hormone glucose-dependent insulinotropic polypeptide (GIP), which facilitates insulin release after a meal K-cells are also glucose responsive, have exocytotic mechanisms, and contain the necessary enzymes for pro-cessing proinsulin to insulin A murine intestinal cell line containing K-cells transfected with human insulin DNA cloned under the control of the GIP promoter produced bio-logically active insulin in response to glucose, and transgenic mice expressing the human proinsulin gene under the GIP promoter were protected from diabetes after treatment with
surro-streptozotocin (Cheung et al., 2000) These results suggest
that K-cells may have great potential as surrogate beta-cells.The strategy of engineering hepatocytes to produce
insulin has been widely studied (Nett et al., 2003)
Hepato-cytes have advantages for engineering as insulin-producing
Trang 19638 C H A P T E R F O R T Y - T H R E E • E N G I N E E R I N G P A N C R E A T I C B E T A - C E L L S
cells because they express components of a glucose-sensing
system somewhat similar to that in pancreatic beta-cells,
such as GLUT2 and glucokinase In addition, there are
several hepatocyte-specifi c gene promoters that respond to
changes in glucose concentrations The L-type pyruvate
kinase promoter (LPK) and Spot14 promoter have been
investigated as regulatory elements for glucose-responsive
insulin production in liver Using a chimeric promoter
com-posed of three copies of the stimulatory glucose-responsive
element from the LPK promoter and an inhibitory
respon-sive element from the insulin-like growth factor–binding
protein-1 basal promoter, the expression of a modifi ed
human proinsulin gene was stimulated by glucose and
inhibited by insulin in hepatocytes Engineering of rat
hepa-toma cells to express insulin under the control of the
glucose-6-phosphatase promoter resulted in the stimulation of
insulin production by glucose and self-limitation by insulin
However, insulin expression by the glucose-6-phosphatase
promoter was low because of negative feedback by the
pro-duced insulin It was recently reported that human
hepa-toma cells transduced with a furin-cleavable human
preproinsulin gene under the control of the GLUT2
pro-moter expressed insulin in response to glucose (Burkhardt
et al., 2005).
A drawback for the regulation of insulin production
by glucose-responsive promoters in hepatocytes is slow as
compared with the rapid release by exocytosis from
beta-cells Because a longer period of time is required for
transcriptional regulation to change the plasma levels of
insulin in response to changes in blood glucose,
hypoglyce-mia may occur Therefore, the development of systems that
mimic insulin secretory dynamics is required Strategies
that utilize synthetic promoters composed of multiple
copies of glucose-responsive elements for the induction of
high levels of insulin expression, insulin-sensitive elements
for feedback regulation, and methods to control of the
half-life of insulin mRNA so that it rapidly degrades may make
it possible to mimic insulin production in a
glucose-responsive fashion in non-beta-cells
Another consideration is that most non-beta-cells do
not have the appropriate endoproteases to convert
proinsu-lin to insuproinsu-lin or secretory granules from which insuproinsu-lin can
be rapidly released in response to physiological stimuli One
approach is the mutation of the proinsulin gene so that it
can be cleaved and converted to insulin by the protease
furin, which is expressed in a wide variety of cells Another
approach is the development of a single-chain insulin
analog, which shows insulin activity without the
require-ment for processing Artifi cially regulated insulin secretion
in non-beta-cells has been tried by expressing insulin as a
fusion protein containing an aggregation domain, which
accumulates in the endoplasmic reticulum and is secreted
when a drug that induces disaggregation is administered
Although engineering somatic non-beta-cells to
pro-duce insulin is a very attractive method, no method has yet
succeeded in imitating normal beta-cells regarding the rapid and tight regulation of glucose within a narrow physi-ological range Improvements, including better control of glucose-responsive transcription of transgenic insulin mRNA and artifi cial secretory systems, provide hope for the potential use of insulin-producing non-beta-cells to cure diabetes
Engineering Stem and Progenitor Cells
An exciting advance in the last few years is the ment of cell therapy strategies using stem cells Stem cells are characterized by the ability to proliferate extensively and differentiate into one or more specialized cell types Both embryonic and adult stem cells have been investigated as alternative sources for the generation of insulin-producing pancreatic islets Although spontaneous differentiation of beta-cells from stem cells can be observed, engineering of stem cells for forced expression of key beta-cell or endocrine differentiation factors should be more effi cient for driving beta-cell differentiation
develop-Engineering Embryonic Stem Cells
In principle, embryonic stem (ES) cells have the tial to generate unlimited quantities of insulin-producing cells ES cells can be expanded indefi nitely in the undiffer-entiated state and differentiated into functional beta-cells
poten-However, generation of fully differentiated beta-cells from
ES cells has been diffi cult and controversial Beta-cell ferentiation from ES cells as determined on the basis of immunohistochemical evidence alone has been questioned, because insulin immunoreactivity can also result from insulin absorption from the medium as well as from genuine beta-cell differentiation Therefore, these types of results should be interpreted with caution
dif-Some promising results have been reported for the differentiation of insulin-producing cells from mouse and human ES cells (reviewed in Bonner-Weir and Weir, 2005;
Jun and Yoon, 2005; Montanya, 2004; Stoffel et al., 2004)
Pancreatic endocrine cells, including insulin-producing cells, could be generated from mouse embryonic stem cells
by a fi ve-step protocol, including the enrichment of positive cells from embryoid bodies, and these cells secreted insulin in response to glucose and other insulin secreta-gogues, such as tolbutamide and carbachol However, these cells could not remit hyperglycemica when transplanted into diabetic mice A modifi ed protocol, in which a phos-phoinositide kinase inhibitor was added to the medium to inhibit cell proliferation, resulted in improved insulin content and glucose-dependent insulin release To enrich insulin-producing cells from mouse ES cells, a neomycin-resistance gene regulated by the insulin promoter was transferred to ES cells, which drove differentiation of insulin-secreting cells, and transplantation of these cells restored normoglycemia in streptozotocin-induced diabetic mice
nestin-In another report, mouse ES cells were transduced with a
Trang 20plasmid containing the Nkx6.1 promoter gene, followed by
a neomycin-resistance gene to select the Nkx6.1-positive
cells, and were differentiated in the presence of exogenous
differentiating factors The selected Nkx6.1-positive cells
coexpressed insulin and Pdx-1, and transplantation of these
cells into streptozotocin-induced diabetic mice resulted in
normoglycemia
Exogenous expression of beta-cell transcription factors
has been used as a strategy to drive the differentiation of
insulin-producing cells from ES cells Overexpression of
Pax4 in mouse ES cells promoted the differentiation
of nestin-positive progenitor and insulin-producing cells,
and these cells secreted insulin in response to glucose
and normalized blood glucose when transplanted into
diabetic mice (Blyszczuk et al., 2003) In the same study,
the expression of Pdx-1 did not have a signifi cant effect
on the differentiation of insulin-producing cells from ES
cells However, another study demonstrated that the
regulated expression of Pdx-1 in a murine ES cell line
by the tet-off system enhanced the expression of insulin
and other beta-cell transcription factors (Miyazaki et al.,
2004)
It was shown that human ES cells can spontaneously
differentiate in vitro into insulin-producing beta-cells,
evi-denced by the secretion of insulin and expression of other
beta-cell markers (Assady et al., 2001) Differentiation of
insulin-expressing cells from human ES cells was promoted
when they were cultured in conditioned medium in the
presence of low glucose and fi broblast growth factor,
fol-lowed by nicotinamide (Segev et al., 2004) A recent report
suggested that human ES cells differentiated into
beta-cell-like clusters when cotransplanted with mouse dorsal
pancreas (Brolen et al., 2005) Although several in vitro
studies suggest the possibility of generating
insulin-expressing cells from human ES cells, differentiation of truly
functional beta-cells from human ES cells has not yet been
reported
Because of their proliferative ability and capacity to
dif-ferentiate in culture, ES cells have received much attention
as a potential source of unlimited quantities of beta-cells for
transplantation therapy for diabetes However, use of ES
cells has ethical concerns, and the mechanisms by which ES
cells differentiate to produce islets and beta-cells are not
well understood Therefore, further studies are needed to
understand the details of the endoderm and beta-cell
differentiation process so that an effective protocol for
differentiating ES cells into insulin-producing cells can be
developed
Engineering Adult Stem and Progenitor Cells
As with ES cells, adult stem cells have the potential to
differentiate into other cell lineages, but they do not bring
the ethical diffi culties associated with ES cells Beta-cell
neogenesis in adults has been reported in animal models of
experimentally induced pancreatic damage, suggesting the
presence of adult stem/progenitor cells These adult stem/progenitor cells could be potential sources for the produc-tion of new insulin-producing cells (reviewed in Jun and Yoon, 2005; Montanya, 2004; Nir and Dor, 2005) Bone marrow, mesenchymal splenocytes, neural stem cells, liver oval stem cells, and pancreatic stem cells have been inves-tigated for their potential to differentiate into insulin-producing cells
A large body of evidence suggests that the adult atic ducts are the main site of beta-cell progenitors Through-out life, the islets of Langerhans turn over slowly, and new small islets are continuously generated by differentiation of
pancre-ductal progenitors (Finegood et al., 1995) It was found that isletlike clusters were generated in vitro from mouse pan-
creatic ducts and ductal tissue–enriched human pancreatic islets In addition, multipotent precursor cells clonally iden-tifi ed from pancreatic islets and ductal populations could differentiate into cells with beta-cell function The expres-sion of the Pdx-1 gene or treatment of ductal cells with Pdx-
1 protein increased the number of insulin-positive cells or induced insulin expression Ectopic expression of neuro-genin 3, a critical factor for the development of the endo-crine pancreas in humans, in pancreatic ductal cells led to their conversion into insulin-expressing cells In addition, treatment of human islets containing both ductal and ascinar cells with a combination of epidermal growth factor and gastrin induced neogenesis of islet beta-cells from the ducts and increased the functional beta-cell mass In addi-tion to ductal cells, exocrine acinar cells and other endo-crine cells can generate beta-cells An alpha-cell line transfected with Pdx-1 expressed insulin when treated with betacellulin It was shown that treatment of rat exocrine pancreatic cells with epidermal growth factor and leukemia inhibitory factor could induce differentiation into insulin-
producing beta-cells (Baeyens et al., 2005) Considerable
evidence suggests that beta-cells in the pancreatic islets can
be dedifferentiated, expanded, and redifferentiated into beta-cells by inducing the epithelial–mesenchymal transi-
tion process (Lechner et al., 2005) Nonendocrine
pancre-atic epithelial cells also have been reported to differentiate
into beta-cells (Hao et al., 2006) These results suggest that
pancreatic stem/progenitor cells are the source of new islets
There is also the possibility of manipulating genitor cells from other organs to transform into the beta-cell phenotype (reviewed in Montanya, 2004; Nir and Dor, 2005) Although there are controversies regarding the differen-tiation of bone marrow–derived stem cells into insulin-producing cells, some successful studies have been reported
stem/pro-In vitro differentiation of mouse bone marrow cells resulted
in the expression of genes related to pancreatic beta-cell development and function These differentiated cells released insulin in response to glucose and reversed hyperglycemia when transplanted into diabetic mice In addition, ectopic expression of key transcription factors of the endocrine
Trang 21640 C H A P T E R F O R T Y - T H R E E • E N G I N E E R I N G P A N C R E A T I C B E T A - C E L L S
pancreas developmental pathway, such as IPF1, HLXB9, and
FOXA2, in combination with conditioned media in human
bone marrow mesenchymal stem cells differentiated them
into insulin-expressing cells (Moriscot et al., 2005).
Because the liver and intestinal epithelium are derived
from gut endoderm, as is the pancreas, the generation of
islets from both developing and adult liver and intestinal
cells has been tried Rat hepatic oval stem cells could
dif-ferentiate into insulin-producing isletlike cells when
cul-tured in a high-glucose environment Fetal human liver
progenitor cells and mouse hepatocytes could differentiate
into insulin-producing cells when engineered to produce
Pdx-1, and transplantation of these cells reversed
hypergly-cemia in mice It was reported that adult human liver cells
engineered to express Pdx-1 produced insulin and secreted
it in a glucose-regulated manner Transplantation of these
engineered cells under the renal capsule of diabetic mice
resulted in prolonged reduction of hyperglycemia (Sapir
et al., 2005) As well, ectopic islet neogenesis in the liver
could be induced by gene therapy with a combination of
NeuroD, a transcription factor downstream of Pdx-1, and
betacellulin, which reversed diabetes in
streptozotocin-treated diabetic mice Expression of Pdx-1 in a rat
entero-cyte cell line in combination with betacellulin treatment or
coexpression of Isl-1 resulted in the expression of insulin
Treatment of developing as well as adult mouse intestinal
cells with GLP-1 induced insulin production, and
transplan-tation of these cells into streptozotocin-induced diabetic
mice remitted diabetes A recent study showed that neural
progenitor cells could generate glucose-responsive,
insulin-producing cells when exposed in vitro to a series of signals
for pancreatic islet development (Hori et al., 2005) These
results suggest that the controlled differentiation of liver or
intestinal cells into insulin-producing cells may provide an
alternative source of beta-cells
The use of adult stem/progenitor cells for generating
beta-cells for transplantation therapy appears to be
promis-ing, although most of the studies have only been done in
animal models Further studies on the mechanisms for the
differentiation of adult stem/progenitor cells into
insulin-producing beta-cells and the characterization of the newly
generated beta-cells are required before these cells can be
considered for clinical application
III ENGINEERING TO IMPROVE
ISLET SURVIVAL
A hurdle to overcome for islet transplantation therapy
is the rejection and autoimmune attack against the
trans-planted beta-cells Immunosuppressive drugs have been
used successfully, but they have many side effects
There-fore, it is desirable to develop drug-free strategies for the
induction of tolerance to transplanted islet or beta-cells A
variety of approaches to protect islet grafts have been
studied, such as bone marrow transplantation, treatment
with anti-T-cell agents, and inhibition of activation of antigen-presenting cells Another approach is engineering islets or beta-cells to express therapeutic genes to improve islet viability and function, such as genes for cytokines, antiapoptotic molecules, antioxidants, immunoregulatory molecules, and growth factors (reviewed in Giannoukakis and Trucco, 2005; Jun and Yoon, 2005; Van Linthout and Madeddu, 2005) (Table 43.1)
With regard to cytokines, introduction of genes for interleukin (IL)-4 or a combination of IL-10 and transform-ing growth factor-β improved islet graft survival by prevent-ing immune attack in mice In addition, islets expressing the p40 subunit of IL-12 could maintain normoglycemia when transplanted into diabetic NOD recipients by decreasing interferon-γ production and increasing transforming growth
Table 43.1 Engineering islets for beta-cell survival
Cytokine expression Erythropoietin
Interleukin (IL)-1 receptor antagonist
IL-4IL-10IL-12p40Transforming growth factor-β
expression Bcl-xL
Dominant-negative MyD88Fas ligand
Flice-like inhibitory protein
IκB kinase inhibitorTumor necrosis factor receptor- immunoglobulin (Ig)
Glutathione peroxidaseHeme oxygenase-1Manganese superoxide dismutaseImmunoregulatory Adenoviral E3 genes
antigen-4-IgDipeptide boronic acidIndoleamine 2,3-dioxygenase
Vascular endothelial growth factor
Trang 22factor-β at the transplantation site Islets engineered to
produce IL-1β receptor antagonist were also found to be
more resistant to rejection Adenoviral-mediated gene
trans-fer of erythropoietin, a cytokine that promotes survival, in
islets resulted in protection of islets from apoptosis in
culture and destruction in vivo.
Expression of antiapoptotic molecules such as Bcl-2,
Bcl-xL, and A20, which inhibit nuclear factor-κB activation,
or an IκB kinase inhibitor was shown to protect from
apop-tosis In addition, the expression of soluble human Fas
ligand, dominant negative MyD88, fl ice-like inhibitory
protein, or tumor necrosis factor receptor-immunoglobulin
(Ig) improved allogeneic islet graft survival A recent study
demonstrated that silencing Fas expression with small
inter-fering RNA in mouse insulinoma cells inhibited
Fas-medi-ated beta-cell apoptosis (Burkhardt et al., 2006).
Pancreatic islets are sensitive to oxidative stress because
they produce relatively low amounts of antioxidant enzymes
Thus, expression of antioxidant molecules such as catalase,
glutathione perioxidase, and manganese superoxide
dis-mutase in islets or insulinoma cells could protect against
oxidative stress and cytokine-induced damage In addition,
expression of heme oxygenase in pancreatic islets protected
against IL-1β-induced islet damage It was also found that
delivery of a c-Jun-terminal kinase inhibitory peptide into
isolated islets by the protein transduction system prevented
apoptosis (Noguchi et al., 2005).
Expression of immunoregulatory molecules that affect
T-cell activation and proliferation have been tried
Expres-sion in islets of cytotoxic T-lymphocyte
antigen-4-immuno-globulin, which down-regulates T-cell activation, or CD40-Ig,
which blocks CD40–CD40 ligand interactions, prolonged
allogenic and xenogeneic graft survival Transplantation
of islets overexpressing indoleamine 2,3-dioxygenase
pro-longed survival in NOD/SCID mice after adoptive transfer
of diabetogenic T-cells, probably by inhibiting T-cell
prolif-eration by the depletion of tryptophan at the
transplanta-tion site Similarly, a proteasome inhibitor, dipeptide boronic
acid, was found to prevent islet allograft rejection by
sup-pressing the proliferation of T-cells Expression of
adenovi-ral E3 transgenes in beta-cells was found to prevent islet
destruction by autoimmune attack through the inhibition of
major histocompatibility complex I expression
With regard to growth factors, adenoviral-mediated
transfer of hepatocyte growth factor resulted in an improved
islet transplant outcome in animal models As well,
expres-sion of insulinlike growth factor-1 in human islets prevented
IL-1βinduced betacell dysfunction and apoptosis Insuffi
-cient revascularization of transplanted islets can deprive
them of oxygen and nutrients, contributing to graft failure
Therefore the expression of vascular endothelial growth
factor, a key angiogenic molecule, enhanced islet
revascu-larization and improved the long-term survival of murine
islets after transplantation into the renal capsule of diabetic
mice
Another strategy to protect islets from immune attack
is microencapsulation of islets within synthetic polymers
(Kizilel et al., 2005) Encapsulation of islets within a
semi-permeable membrane, such as nate, blocks the passage of larger cells but allows the passage
alginatepolyllysinealgiof small molecules, thus conferring protection from auto
-i mmune attack However, th-is method has l-im-itat-ions for the long-term survival of islets within the microcapsules because of the lack of biocompatibility, ischemia, and limited protection from cytokine-induced damage To over-come these limitations, a bioartifi cial pancreas has been developed, in which blood fl ows through artifi cial vessels in close proximity to insulin-producing cells
A variety of approaches for engineering islets or cells for improved islet graft survival and escape from immune rejection have been successful in animal models However, the effi cacy of these approaches in human dia-betic patients remains to be determined
beta-IV VECTORS FOR ENGINEERING ISLETS AND BETA-CELLS
The cells of the pancreas divide very slowly; therefore gene transfer vehicles that can transduce quiescent cells have been used for the delivery of transgenes, such as nonviral plasmids and vectors based on lentivirus, adeno-virus, helper-dependent adenovirus, adeno-associated virus (AAV), and herpes simplex virus In addition, protein trans-duction using the cell penetrating peptide from HIV-1 trans-
acting protein (reviewed in Becker-Hapak et al., 2001) has
been successfully used to engineer islets (Table 43.2) However, the choice of vector needs to be carefully made so that the vector itself does not affect islet function or viability
Nonviral methods are considered safe, cost effective, and simple to use and do not induce an immune response, but they generally have a lower gene transfer effi cacy as compared to viral-mediated gene transfer (reviewed in Nishikawa and Huang, 2001) Nonviral methods for transferring genetic material include the direct injection
of DNA, either naked or enclosed in a liposome, poration, and the gene gun method Cationic lipid and polymer-based plasmid delivery has been used to trans-duce islets, and the expression of cytotoxic T-lymphocyte antigen-4 by biolistically transfected islets improved graft survival
electro-Viral vectors (reviewed in Walther and Stein, 2000) have been widely used as a method of gene transfer to engineer islets and beta-cell surrogates Retroviral vectors derived from Moloney murine leukemia virus can carry a gene effi -ciently and integrate it in a stable manner within the host chromosomal DNA, facilitating long-term expression of the gene For immortalization of human islets, retroviral vectors expressing oncogenes or telomerase genes have been used
(Narushima et al., 2005) Although most retroviral vectors
Trang 23642 C H A P T E R F O R T Y - T H R E E • E N G I N E E R I N G P A N C R E A T I C B E T A - C E L L S
only infect proliferating cells, the lentivirus genus of
retro-viruses, which includes the human immunodefi ciency virus,
has all the advantages of Moloney murine leukemia virus–
derived retroviral vectors and can infect nondividing as well
as dividing cells Lentiviral vectors have been successfully
used to transduce islets with marker proteins (Okitsu et al.,
2003)
The adenoviral vector can harbor up to 30 Kb of foreign
DNA and can transduce nondividing cells with high effi
-ciency In addition, a relatively high titer of virus, about 1012
plaque-forming units/mL, can be produced The transferred
genes are not integrated into the host genome, but remain
as nonreplicating extrachromosomal DNA within the
nucleus Although there is no risk of alteration in cellular
genotype by insertional mutation, the duration of gene
expression may be short, and a strong cellular immune
response to the viral proteins and, in some cases, to the
transgene may be induced Adenoviral vectors have been
widely used to transduce islets for proof-of-concept
experi-ments in vitro and in vivo Although adenoviral vectors are
toxic because of de novo synthesized viral proteins, islet
viability and functional characteristics were not affected
when transduced in vitro However, transduction of islets
with a high dose of recombinant adenovirus (500 MOI) markedly reduced glucose-stimulated insulin secretion, suggesting that an optimal dose is required to result in effi -cient transduction without compromising islet function In general, adenoviral vectors result in transient transgene expression; however, long-term (20-week) expression of the transgene was observed in islets transduced with β-galacto-sidase and transplanted into syngeneic diabetic mice It was reported that double-genetic modifi cation of the adeno-virus fi ber with RGD polylysine motifs signifi cantly reduced toxicity, infl ammation, and immune responses (Contreras
et al., 2003).
A new generation of adenovirus vectors has been oped that are completely devoid of all viral protein–coding sequences and are therefore less immunogenic and less toxic Although these gutless viruses require the presence of
devel-a helper virus for replicdevel-ation, contdevel-amindevel-ation by the helper virus can be avoided by genetically engineering a condi-tional defect in the packaging domain of the helper virus or
fl anking the packing signal with loxP expression sites and encoding Cre recombinase in the supporting cell line In
Table 43.2 Vectors used for islet and beta-cell engineering
Less toxic compared with viral vectors Transient expression
Random integration into host chromosomal DNA
Infects dividing and nondividing cells Limited insertion capacity (8 Kb)
DNA
Infects dividing and nondividing cells Short-term expression
compared with adenoviral vectorLarge insertion capacity
Long-term expressionInfects dividing and nondividing cells
Transduces many cell types
Trang 24addition, the gutless vectors are known to have a prolonged
expression of the transgene However, there is no report
about islet engineering using these vectors
AAVs are nonpathogenic, replication-defective
parvo-viruses that can infect both dividing and nondividing cells
AAVs generally have low immunogenicity; however, the
generation of neutralizing antibodies may limit
readminis-tration This problem can be overcome by selective capsid
modifi cation of AAV to evade recognition by preexisting
anti-bodies or by direct administration of AAV to the target tissue
The recombinant AAV vector integrates randomly into the
host chromosome or may stay in the episomal state There
is a limitation in the size of the DNA that can be inserted (a
maximum of 4.8 Kb); however, larger inserts can be split over
two vectors and delivered simultaneously, because AAVs
tend to form concatemers, although the effi ciency of
duction is often reduced (Young et al., 2006) Effi cient
trans-duction of islets was achieved using a high dose of AAVs with
an improved recombinant AAV purifi cation method, which
improved infectious titers and yield Transduction of islets
with AAV5 is more effi cient than with AAV2, due to the low
number of receptors for AAV2 on islet cells AAV1 was found
to be the most effi cient serotype in transducing murine islets
(Loiler et al., 2003) However, it was recently demonstrated
that intact human and murine islets could be effi ciently
transduced with a double-stranded AAV2-based vector, and
the transduced murine islets showed normal glucose
respon-siveness and viability (Rehman et al., 2005).
Herpes simplex virus type 1 (HSV-1) has also been used
as a viral vector Based on the persistence of latent herpes
virus after infection, HSV-1 is attractive for its effi cient
infec-tivity in a wide range of target cells and its ability to infect
both dividing and nondividing cells, including islets
Trans-fection of human islets with Bcl-2 protected beta-cells from
cytokine-induced damage However, the transduction may
be unstable, and potential health risks of this vector remain
to be determined
Protein transduction is an emerging technology to
deliver therapeutic proteins into cells as an alternative to
gene therapy This method uses peptides that can penetrate the cell membrane, such as antennapedia peptide, the HSV VP22 protein, and human immunodefi ciency virus TAT protein transduction domain The therapeutic molecule
is linked to the penetrating peptide as a fusion protein, which is then used to transduce the cell (Becker-Hapak
et al., 2001) The protein transduction method is not
im-munogenic and can transduce a variety of cell types, but it has a short half-life Delivery of antiapoptotic proteins such as Bcl-xL or anti-oxidant enzymes such as copper-zinc superoxide dismutase and heme oxygenase by protein transduction in human and rodent islets effi ciently trans-duced the islets and improved their viability without
affecting islet function (Embury et al., 2001; Mendoza et al.,
2005)
V CONCLUSION
Engineering beta-cell lines and non-beta-cells, entiating embryonic and adult stem cells, and transdiffer-entiating non-beta-cells have been studied as methods to provide new beta-cells for cell therapy for diabetes Expan-sion of functional beta-cells by generation of reversibly immortalized human beta-cell lines has been reported, but the techniques have not been clinically proven Generation
differ-of insulin-producing cells from non-beta-cells is an tive method, but it has yet to achieve tight regulation of glucose-responsive insulin secretion Differentiation of insulin-producing cells from ES cells and adult stem/progenitor cells is also a promising alternative to produce beta-cells; however, a better understanding of the mecha-nisms for the differentiation of beta-cells is needed to develop a successful strategy to engineer beta-cells from stem cells Engineering of islets and beta-cells to improve the survival of islet transplants has also been investigated Although much progress has been made, engineered beta-cells need to be carefully analyzed for true beta-cell function and possible tumorigenicity It is hoped that continued research on beta-cell engineering will offer a potential cure for diabetes in the future
attrac-VI REFERENCES
Adorini, L., Gregori, S., and Harrison, L C (2002) Understanding
autoimmune diabetes: insights from mouse models Trends Mol Med
8, 31–38.
Assady, S., Maor, G., Amit, M., Itskovitz-Eldor, J., Skorecki, K L., and
Tzukerman, M (2001) Insulin production by human embryonic stem
cells Diabetes 50, 1691–1697.
Baeyens, L., De Breuck, S., Lardon, J., Mfopou, J K., Rooman, I., and
Bouwens, L (2005) In vitro generation of insulin-producing beta cells
from adult exocrine pancreatic cells Diabetologia 48, 49–57.
Beattie, G M., Itkin-Ansari, P., Cirulli, V., Leibowitz, G., Lopez, A D.,
Bossie, S., Mally, M I., Levine, F., and Hayek, A (1999) Sustained
proliferation of PDX-1+ cells derived from human islets Diabetes 48,
1013–1019.
Becker-Hapak, M., McAllister, S S., and Dowdy, S F (2001)
TAT-mediated protein transduction into mammalian cells Methods 24,
247–256.
Blyszczuk, P., Czyz, J., Kania, G., Wagner, M., Roll, U., St-Onge, L., and Wobus, A M (2003) Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-
producing cells Proc Natl Acad Sci U.S.A 100, 998–1003.
Bonner-Weir, S., and Weir, G C (2005) New sources of pancreatic
beta-cells Nat Biotechnol 23, 857–861.
Brolen, G K., Heins, N., Edsbagge, J., and Semb, H (2005) Signals from the embryonic mouse pancreas induce differentiation of human
embryonic stem cells into insulin-producing beta-cell-like cells
Diabe-tes 54, 2867–2874.
Trang 25644 C H A P T E R F O R T Y - T H R E E • E N G I N E E R I N G P A N C R E A T I C B E T A - C E L L S
Burkhardt, B R., Parker, M J., Zhang, Y C., Song, S., Wasserfall, C H.,
and Atkinson, M A (2005) Glucose transporter-2 (GLUT2) promoter–
mediated transgenic insulin production reduces hyperglycemia in
dia-betic mice FEBS Lett 579, 5759–5764.
Burkhardt, B R., Lyle, R., Qian, K., Arnold, A S., Cheng, H., Atkinson,
M A., and Zhang, Y C (2006) Effi cient delivery of siRNA into
cytokine-stimulated insulinoma cells silences Fas expression and inhibits
Fas-mediated apoptosis FEBS Lett 580, 553–560.
Cheung, A T., Dayanandan, B., Lewis, J T., Korbutt, G S., Rajotte, R V.,
Bryer-Ash, M., Boylan, M O., Wolfe, M M., and Kieffer, T J (2000)
Glucose-dependent insulin release from genetically engineered K cells
Science 290, 1959–1962.
Contreras, J L., Wu, H., Smyth, C A., Eckstein, C P., Young, C J., Seki,
T., Bilbao, G., Curiel, D T., and Eckhoff, D E (2003) Double genetic
modifi cation of adenovirus fi ber with RGD polylysine motifs signifi
-cantly enhances gene transfer to isolated human pancreatic islets
Transplantation 76, 252–261.
de la Tour, D., Halvorsen, T., Demeterco, C., Tyrberg, B., Itkin-Ansari, P.,
Loy, M., Yoo, S J., Hao, E., Bossie, S., and Levine, F (2001) Beta-cell
dif-ferentiation from a human pancreatic cell line in vitro and in vivo Mol
Endocrinol 15, 476–483.
Embury, J., Klein, D., Pileggi, A., Ribeiro, M., Jayaraman, S., Molano,
R D., Fraker, C., Kenyon, N., Ricordi, C., Inverardi, L., et al (2001)
Pro-teins linked to a protein transduction domain effi ciently transduce
pancreatic islets Diabetes 50, 1706–1713.
Finegood, D T., Scaglia, L., and Bonner-Weir, S (1995) Dynamics of
beta-cell mass in the growing rat pancreas Estimation with a simple
mathematical model Diabetes 44, 249–256.
Giannoukakis, N., and Trucco, M (2005) Gene therapy for type 1
dia-betes Am J Ther 12, 512–528.
Halvorsen, T L., Leibowitz, G., and Levine, F (1999) Telomerase activity
is suffi cient to allow transformed cells to escape from crisis Mol Cell
Biol 19, 1864–1870.
Hao, E., Tyrberg, B., Itkin-Ansari, P., Lakey, J R., Geron, I., Monosov,
E Z., Barcova, M., Mercola, M., and Levine, F (2006) Beta-cell
differen-tiation from nonendocrine epithelial cells of the adult human pancreas
Nat Med 12, 310–316.
Hatipoglu, B., Benedetti, E., and Oberholzer, J (2005) Islet
trans-plantation: current status and future directions Curr Diab Rep 5,
311–316.
Hori, Y., Gu, X., Xie, X., and Kim, S K (2005) Differentiation of
insulin-producing cells from human neural progenitor cells PLoS Med 2,
e103.
Islam, M S., Rahman, S A., Mirzaei, Z., and Islam, K B (2005)
Engineered beta-cells secreting dipeptidyl peptidase IV–resistant
glucagon-like peptide-1 show enhanced glucose responsiveness Life
Sci 76, 1239–1248.
Jun, H S., and Yoon, J W (2005) Approaches for the cure of type 1
dia-betes by cellular and gene therapy Curr Gene Ther 5, 249–262.
Kizilel, S., Garfi nkel, M., and Opara, E (2005) The bioartifi cial
pancreas: progress and challenges Diabetes Technol Ther 7, 968–985.
Lechner, A., Nolan, A L., Blacken, R A., and Habener, J F (2005)
Redif-ferentiation of insulin-secreting cells after in vitro expansion of adult
human pancreatic islet tissue Biochem Biophys Res Commun 327,
581–588.
LeRoith, D (2002) Beta-cell dysfunction and insulin resistance in type
2 diabetes: role of metabolic and genetic abnormalities Am J Med
113(Suppl 6A), 3S–11S.
Loiler, S A., Conlon, T J., Song, S., Tang, Q., Warrington, K H., Agarwal,
A., Kapturczak, M., Li, C., Ricordi, C., Atkinson, M A., et al (2003)
Targeting recombinant adeno-associated virus vectors to enhance
gene transfer to pancreatic islets and liver Gene Ther 10, 1551–
1558.
Mendoza, V., Klein, D., Ichii, H., Ribeiro, M M., Ricordi, C., Hankeln, T., Burmester, T., and Pastori, R L (2005) Protection of islets in culture by delivery of oxygen-binding neuroglobin via protein transduction
Transplant Proc 37, 237–240.
Milo-Landesman, D., Surana, M., Berkovich, I., Compagni, A., Christofori, G., Fleischer, N., and Efrat, S (2001) Correction of hyper- glycemia in diabetic mice transplanted with reversibly immortalized
pancreatic beta cells controlled by the tet-on regulatory system Cell
Transplant 10, 645–650.
Miyazaki, S., Yamato, E., and Miyazaki, J (2004) Regulated expression
of pdx-1 promotes in vitro differentiation of insulin-producing cells
from embryonic stem cells Diabetes 53, 1030–1037.
Montanya, E (2004) Islet- and stem-cell-based tissue engineering in
diabetes Curr Opin Biotechnol 15, 435–440.
Moriscot, C., de Fraipont, F., Richard, M J., Marchand, M., Savatier, P., Bosco, D., Favrot, M., and Benhamou, P Y (2005) Human bone marrow mesenchymal stem cells can express insulin and key transcription factors of the endocrine pancreas developmental pathway upon genetic
and/or microenvironmental manipulation in vitro Stem Cells 23,
594–603.
Narushima, M., Kobayashi, N., Okitsu, T., Tanaka, Y., Li, S A., Chen, Y.,
Miki, A., Tanaka, K., Nakaji, S., Takei, K., et al (2005) A human beta-cell line for transplantation therapy to control type 1 diabetes Nat Biotech-
nol 23, 1274–1282.
Nett, P C., Sollinger, H W., and Alam, T (2003) Hepatic insulin gene
therapy in insulin-dependent diabetes mellitus Am J Transplant 3,
1197–1203.
Nir, T., and Dor, Y (2005) How to make pancreatic beta cells —
prospects for cell therapy in diabetes Curr Opin Biotechnol 16, 524–
529.
Nishikawa, M., and Huang, L (2001) Nonviral vectors in the new
mil-lennium: delivery barriers in gene transfer Hum Gene Ther 12,
861–870.
Noguchi, H., Nakai, Y., Matsumoto, S., Kawaguchi, M., Ueda, M., Okitsu,
T., Iwanaga, Y., Yonekawa, Y., Nagata, H., Minami, K., et al (2005)
Cell-permeable peptide of JNK inhibitor prevents islet apoptosis
immedi-ately after isolation and improves islet graft function Am J Transplant
5, 1848–1855.
Okitsu, T., Kobayashi, N., Totsugawa, T., Maruyama, M., Noguchi, H., Watanabe, T., Matsumura, T., Fujiwara, T., and Tanaka, N (2003) Len- tiviral vector–mediated gene delivery into nondividing isolated islet
cells Transplant Proc 35, 483.
Rehman, K K., Wang, Z., Bottino, R., Balamurugan, A N., Trucco, M.,
Li, J., Xiao, X., and Robbins, P D (2005) Effi cient gene delivery to human and rodent islets with double-stranded (ds) AAV-based vectors
Natl Acad Sci U.S.A 102, 7964–7969.
Segev, H., Fishman, B., Ziskind, A., Shulman, M., and Itskovitz-Eldor, J
(2004) Differentiation of human embryonic stem cells into
insulin-producing clusters Stem Cells 22, 265–274.
Trang 26Stoffel, M., Vallier, L., and Pedersen, R A (2004) Navigating the pathway
from embryonic stem cells to beta cells Semin Cell Dev Biol 15,
327–336.
Van Linthout, S., and Madeddu, P (2005) Ex vivo gene transfer for
improvement of transplanted pancreatic islet viability and function
Curr Pharm Des 11, 2927–2940.
Walther, W., and Stein, U (2000) Viral vectors for gene transfer: a review
of their use in the treatment of human diseases Drugs 60, 249–271.
Wu, L., Nicholson, W., Wu, C Y., Xu, M., McGaha, A., Shiota, M., and
Powers, A C (2003) Engineering physiologically regulated insulin
secretion in non-beta cells by expressing glucagon-like peptide 1
recep-tor Gene Ther 10, 1712–1720.
Xu, R., Li, H., Tse, L Y., Kung, H F., Lu, H., and Lam, K S (2003) Diabetes
gene therapy: potential and challenges Curr Gene Ther 3, 65–82.
Yoon, J W., and Jun, H S (2002) Recent advances in insulin gene
therapy for type 1 diabetes Trends Mol Med 8, 62–68.
Yoon, J W., and Jun, H S (2005) Autoimmune destruction of pancreatic
beta cells Am J Ther 12, 580–591.
Young, L S., Searle, P F., Onion, D., and Mautner, V (2006) Viral gene
therapy strategies: from basic science to clinical application J Pathol
208, 299–318.
Trang 27I Introdution
II Structure and Morphology of the Thymus
III In Vitro T-Cell Differentiation
IV Thymus Organogenesis
V Summary
VI Acknowledgments VII References
Thymus and Parathyroid
Organogenesis
Craig Scott Nowell, Ellen Richie, Nancy Ruth Manley, and Catherine Clare Blackburn
Principles of Tissue Engineering, 3 rd Edition
ed by Lanza, Langer, and Vacanti
Copyright © 2007, Elsevier, Inc.
All rights reserved.
I INTRODUTION
The thymus is the principal site of T-cell development
and therefore is of central importance within the immune
system: Congenital athymia results in profound
immunode-fi ciency (Flanagan, 1966; Dodson et al., 1969; J Frank et al.,
1999), while perturbed thymic function can lead to
autoim-munity Although highly active in early life, the thymus
undergoes premature involution, such that de novo T-cell
development diminishes signifi cantly with age This has
implications for immune function in the aging population
and in clinical procedures such as bone marrow and solid
organ transplantation, where thymic function is required
for T-cell reconstitution and/or tolerance induction Interest
therefore exists in enhancing immune reconstitution
through regenerative or cell therapies for boosting thymus
activity in vivo or for providing customized in
vitro–gener-ated T-cell repertoires for adoptive transfer The success of
such strategies is likely to depend on a detailed knowledge
of the mechanisms regulating thymus development and
homeostasis Here, we review current understanding of
cel-lular and molecular regulation of thymus organogenesis,
focusing on the epithelial component of the thymic stroma,
which provides many of the specialist functions required
to mediate T-cell differentiation and T-cell repertoire
mesenchymal cells, bone marrow (BM)–derived cells, culature and the uniquely specialized thymic epithelium
vas-(Boyd et al., 1993).
The mature thymus is encapsulated and lobulated and contains three principal histologically defi ned regions: the cortex, the medulla and the subcapsule (Fig 44.1) The capsule and trabeculae consist of a thick layer of connective tissue and are separated from the cortex by a thin layer of
simple epithelium, the subcapsule (Boyd et al., 1993) The
cortex and medulla each contain open networks of lial cells, which are densely packed with thymocytes (Van
epithe-Vliet et al., 1985; Boyd et al., 1993; van Ewijk et al., 1994), and
each of these regions contains several different cally and phenotypically distinct epithelial subtypes (see later) The outer cortex also contains fi broblasts, and the organ as a whole is heavily vascularized BM-derived stromal cells are found in both compartments, macrophages being distributed throughout the organ, while thymic dendritic
Trang 28cells — which are required for imposition of tolerance on
the emerging T-cell repertoire — are found predominantly
at the corticomedullary junction and in the medulla itself
(Boyd et al., 1993).
Thymus structure is intimately linked to its principal
function, to support T-cell development This encompasses
the linked processes of T-cell differentiation and T-cell
rep-ertoire selection, which together ensure that the peripheral
T-cell repertoire is populated predominantly by T-cells that
have propensity to bind antigen in the context of self-major
histocompatibility antigens (MHC) but that do not bind
self-peptides T-cell development has been extensively
reviewed elsewhere (Zuniga-Pfl ucker and Lenardo, 1996;
Petrie, 2002; Rothenberg and Dionne, 2002) and is not
dis-cussed in detail herein In brief, hematopoietic progenitors
enter the postnatal thymus at the corticomedullary
junc-tion, and subsequent T-cell development is then regulated
such that thymocytes at different stages of development are
found in different intrathymic locations T-cell
differentia-tion from the earliest postcolonizadifferentia-tion stages of thymocyte
maturation [termed triple negative (TN) cells because they
do not express CD3 or the coreceptors CD4 and CD8]
through to the CD4+CD8+ double positive (DP) stage occurs
in the cortex, and the cortex itself can be subdivided into four regions based on the localization of different thymo-cyte populations Thus, zone 1 contains the colonizing population of hematopoietic progenitor cells; these early thymocytes undergo proliferative expansion in zone 2; T-cell lineage commitment is completed in zone 3; and in zone 4, thymocytes differentiate to the DP stage of develop-ment, characterized by expression of both CD4 and CD8
coreceptors (Lind et al., 2001; Porritt et al., 2004) DP cells
are then screened for their propensity to recognize
self-MHC, a process termed positive selection, and those selected
to mature into CD4+ or CD8+ single positive (SP) cells migrate
into the medulla (Kurobe et al., 2006) Central tolerance is
established by deletion of self-reactive thymocytes at the
DP–SP transition, in a process termed negative selection (Baldwin et al., 2005), and is thought to occur principally
at the corticomedullary junction and in the medulla SP
FIG 44.1. Histology of the postnatal thymus The postnatal thymus is surrounded by a capsule consisting of mesenchymal cells and connective tissue
that penetrates into the thymus at regular intervals to form trabeculae Underlying the capsule and trabeculae is the subcapsular epithelium, consisting of
a layer of simple epithelium, which overlies the outer cortex The cortex is populated with cortical thymic epithelial cells (cTEC), macrophages, and developing
thymocytes at the triple negative (TN) and double positive (DP) stages of development Thymocytes enter the thymus at the corticomedullary junction (CMJ)
via the vasculature and migrate through the cortex to the subcapsule as they differentiate The cortex can be divided into four zones based on the
differentia-tion status of thymocytes that reside within it Thus, zone 1 contains the most immature TN1 thymocytes and zone 4 contains thymocytes undergoing the
TN4–DP transition DP thymocytes are then screened for propensity to recognize self-MHC, a process termed positive selection, and those selected to mature
into CD4+ or CD8+ single positive (SP) cells migrate into the medulla, where they undergo the fi nal stages of maturation before being exported to the periphery
Central tolerance is established by deletion of self-reactive thymocytes in a process termed negative selection, and it is thought to occur principally at the
corticomedullary junction and in the medulla: Negative selection is mediated by both thymic dendritic cells and medullary TECs, which supply self-peptides
to medullary dendritic cells (DCs) in a process termed cross-presentation Medullary TECs are also required for the generation of CD4+CD25+ T regulatory
(Treg) cells and natural killer T-cells, both of which actively repress self-reactive T-cells
Trang 29I I I I N V I T R O T - C E L L D I F F E R E N T I A T I O N • 649
thymocytes proliferate and undergo the fi nal stages of
T-cell maturation in the medulla before exiting into the
peri-pheral immune system The outward migration of
thymo-cytes, from the corticomedullary junction to the outer
cortex, is regulated by chemokines (Plotkin et al., 2003), as
is the migration of positively selected cells from the cortex
into the medulla (Ueno et al., 2004; Kurobe et al., 2006).
Thymic Epithelial Cells
The thymic epithelium (TE) can be usefully classifi ed
into three broad subtypes: subcapsular/subtrabecular,
cor-tical and medullary thymic epithelial cells Within these
subdivisions, ultrastructural and immunohistochemical
analyses have revealed at least six different subsets (van de
Wijngaert et al., 1983), assigned as “clusters of thymic
epi-thelial staining” (CTES) types I, II, III, IIIB, IIIC, and IV
(Brekelmans and van Ewijk, 1990) based on different mAb
staining profi les
Thus, subcapsular/subtrabecular epithelium consists
of type 1 epithelial cells in a simple epithelial layer These
cells are MHC Class II negative (Boyd et al., 1992) and are
reactive to CTES II mAbs (Godfrey et al., 1990) The
outer-most cortical subpopulation comprises type II epithelial
cells, characterized by their pale appearance in
electronmi-crographs (van de Wijngaert et al., 1984) Immediately
adja-cent to the type II epithelia are type III thymic epithelial
cells, which show intermediate electron lucency (van de
Wijngaert et al., 1984), while the innermost cTEC are type IV
cells, which have high electron lucency and
oval/spindle-shaped nuclei (van de Wijngaert et al., 1984) Ultrastructural
analysis has also revealed large complexes of type II and
type III cells and developing thymocytes (van de Wijngaert
et al., 1984), termed thymic nurse cells (TNC) (Wekerle and
Ketelsen, 1980a, 1980b) No mAbs are currently known to
identify individual subpopulations corresponding to the
types II–IV cells just described; however, all cTEC stain with
CTES III mAbs (van de Wijngaert et al., 1984), and types II
and III cells are strongly MHC Class II positive (Boyd et al.,
1993) Medullary TEC (mTEC) predominantly express
determinants reactive to CTES II and IV mAbs, with type
III cells also identifi ed by ultrastructural analysis (van de
Wijngaert et al., 1984) In addition, type V epithelia,
classi-fi ed as undifferentiated cells, exist in small isolated clusters
at the corticomedullary junction (van de Wijngaert et al.,
1984), along with type VI cells, which have been proposed
to be precursors of differentiated epithelial cells (von
Gaudecker et al., 1986) All mTEC express MHC Class I,
while MHC Class II expression is variable (Jenkinson
et al., 1981; Farr and Nakane, 1983; Bofi ll et al., 1985; Surh
et al., 1992).
The different thymic epithelial subpopulations are also
defi ned by differential expression of cytokeratins (K) Two
cortical populations have been identifi ed: a predominant
K5−K14−K8+K18+ subset and a minor subset also consisting
of K5+K14−K8+K18+ cells (Klug et al., 1998) Most mTEC
display a K5+K14+K8−K18− phenotype (Klug et al., 1998) and
also express the antigen reactive to mAb MTS10 (Godfrey
et al., 1990), and a minor K5−K14−K8+K18+ MTS10−
medul-lary subset is also present (Klug et al., 1998).
While precise functions are not ascribed to all of these TEC subpopulations, the clear functional dichotomy between the cortical and medullary compartments is refl ected in functional differences between the cortical and medullary thymic epithelial cell types Notably, cortical thymic epithelial cells (cTEC) are believed to express a ligand
required for positive selection (Anderson et al., 1994), while
a subset of medullary thymic epithelial cells (mTEC) expresses AIRE1 (AIRE1 positively regulates expression of a cohort of tissue or developmentally restricted genes that play an essential role in the induction of central tolerance)
(M S Anderson et al., 2002) This high level of phenotypic
and functional heterogeneity presents a signifi cant lenge for attempts to support full T-cell development,
chal-including proper repertoire selection in vitro, and is also
pertinent to cell replacement or regenerative strategies for
enhancing thymus activity in vivo.
III IN VITRO T-CELL DIFFERENTIATION
The ability to generate T-cells in culture is widely used
as a tool for investigating the regulation of T-cell
differentia-tion (Hare et al., 1999) and is also of interest for clinical and
pharmaceutical purposes Several methodologies exist that
are based on the use of ex vivo thymic tissue Thus, fetal thymic organ culture (FTOC) utilizes ex vivo thymic lobes
usually derived from E15.5–E16.5 mouse embryos or trimester human fetuses to support the differentiation of T-cell progenitors from endogeneous or exogenous sources
second-(Jenkinson and Owen, 1990; Yeoman et al., 1993; Barcena
et al., 1994; Plum et al., 1994; Cumano et al., 1996)
The technique of reaggregate fetal thymic organ culture (RFTOC), in which defi ned TEC subpopulations are ob-tained by cell purifi cation techniques, reaggregated with
fi broblasts and defi ned lymphocyte populations, and then
cultured further in vitro, was developed as an extension
of FTOC and has proved invaluable for assessing the role
of individual stromal components during specifi c stages of
T-cell maturation (Jenkinson et al., 1992; G Anderson et al.,
1994) In addition, this approach has recently been adapted for testing the potency of different fetal and adult TEC
subpopulations (Bennett et al., 2002; Gill et al., 2002; Rossi
et al., 2006).
Recently, it has been demonstrated that a
tantalum-coated carbon matrix can be used to generate an in vitro
thymic organoid when seeded with ex vivo murine thymic
stromal cells (Poznansky et al., 2000) When these structures
were cocultured with human CD34+ haematopoietic genitors, effi cient generation of mature CD4 and CD8 SP T-cells was observed after 14 days The T-cells generated in this system were functional, as demonstrated by their prolifera-tive response to mitogenic stimuli, and demonstrated a diverse TCR repertoire comparable to that of peripheral
pro-blood T-cells (Poznansky et al., 2000) These fi ndings
Trang 30dem-onstrate that the utilization of three-dimensional matrices
in conjunction with thymic stromal cells can provide an
effi cient and reproducible method of in vitro T-cell
genera-tion However, this approach currently relies on seeding
with ex vivo thymus tissue and therefore is not highly
scal-able in its present form
In vitro T-cell differentiation has also been investigated
using a derivative of the BM stromal cell line OP-9, which
expresses the Notch ligand Delta-like 1 (OP-9 DL-1) Recent
studies demonstrate that OP-9 DL-1 monolayers can support
the generation of CD4+CD8+ DP thymocytes from mouse
fetal liver–, adult bone marrow–, or ES cell–derived
hema-topoietic progenitors (Schmitt and Zuniga-Pfl ucker, 2002;
Schmitt et al., 2004) Small numbers of CD8+ SP T-cells were
also produced, although CD4+ SP T-cells were largely absent
This system was recently shown to support T-cell
develop-ment from human cord blood– and human bone marrow–
derived CD34+ cells (De Smedt et al., 2002; La Motte-Mohs
et al., 2005) However, although it has been proposed that
this system presents a scalable means of supporting in vitro
T-cell differentiation (Lehar and Bevan, 2002), it remains
unclear to what extent the T-cells generated on OP9-DL1
cells undergo positive and negative selection In an
interest-ing alternative to generation of mature T-cells, this system
has recently been used as a means of expanding a CD4−CD8−
DN precursor thymocyte population, which resulted in
improved T-cell reconstitution after adoptive transfer of
these DN cells in a mouse model of hematopoietic stem cell
transplantation (Zakrzewski et al., 2006) In addition, similar
to RFTOC, the OP-9 DL-1 system is a powerful experimental
tool for examining aspects of T-cell differentiation (Porritt et
al., 2004).
T-cell differentiation in vitro has also been achieved
using preparations of cells derived from human skin in
con-junction with a tantalum-coated carbon matrix (Clark et al.,
2005) In this system, cutaneous keratinocytes and fi
bro-blasts were seeded onto the matrix and supplied with human
CD34+ hematopoietic progenitor cells After a period of
three to four weeks, CD3+ T-cells were produced that
exhib-ited functional maturity and were tolerant to self-MHC as
assessed by the mixed lymphocyte reaction Gene
expres-sion analysis demonstrated that the cells used to seed the
matrix expressed transcription factors associated with TEC
function, such as Foxn1, Aire, and Hoxa3, although at much
lower levels than in the normal thymus However, the effi
-ciency of thymopoiesis in this system was low, with a
rela-tively low number of mature T-cells generated despite the
addition of a cocktail of prolymphopoietic cytokines
IV THYMUS ORGANOGENESIS
Cellular Regulation of
Early Thymus Organogenesis
The thymus arises in the pharyngeal region of the
developing embryo, in a common primordium with the
parathyroid gland This common primordium develops from the third pharyngeal pouch (3PP), one of a series of bilateral outpocketings of pharyngeal endoderm, termed
the pharyngeal pouches, which form sequentially in a
rostral-to-caudal manner
In the mouse, outgrowth of the 3PP occurs from
approx-imately E9.0 (Gordon et al., 2004) At this stage, the
epithe-lium of the 3PP consists of a single layer of columnar epithelium surrounded by a condensing population of neural crest cells (NCC) that will eventually form the capsule
(Le Lievre and Le Douarin, 1975; Jiang et al., 2000) Overt
thymus organogenesis is evident from between E10.5 and E11.0, at which stage the epithelium begins to proliferate,
assuming a stratifi ed organization (Itoi et al., 2001)
Follow-ing this, at E12.5, the primordia separate from the pharynx and begin to resolve into discrete thymus and parathyroid organs The thymus primordium subsequently migrates to its fi nal anatomical location at the midline, while the para-thyroid primodium associates with the lateral margins of the thyroid (Manley and Capecchi, 1995, 1998) In the case of the thymus at least, this migration is active and follows the
path of the carotid artery and vagus nerve (Su et al., 2001).
Within the common primordium, the prospective thymus is located in the ventral domain of the third pouch and the prospective parathyroid in the dorsal aspect Pat-terning of these prospective organ domains appears to occur early in organogenesis, for the parathyroid domain
is delineated by the transcription factor Gcm2 as early as E9.5 Possible mechanisms regulating the establishment/
maintenance of these domains are discussed later
The mesenchymal capsule surrounding the thymus mordium is derived from the migratory neural crest, a tran-sient population formed between the neural tube and the surface ectoderm In the mouse, NCC migrate into the pha-ryngeal region from E9 Elegant chick-quail chimera studies provided the fi rst evidence that NCC are the source of mes-enchymal cells in the thymus (Le Lievre and Le Douarin, 1975), and this was recently confi rmed in the mouse by heri-
pri-table genetic labeling in vivo (Jiang et al., 2000).
Colonization of the mouse thymus with hematopoietic progenitor cells occurs around E11.5 (Owen and Ritter, 1969;
Cordier and Haumont, 1980; Jotereau et al., 1987) Because
vascularization has not occurred by this stage, the fi rst nizing cells migrate through the perithymic mesenchyme into the thymic epithelium These cells have been reported
colo-to exhibit comparatively low T-cell progenicolo-tor activity, while
a second colonizing wave, which arrives between E12 and E14, appears to display much higher levels of T-cell potential
upon in vivo transfer (Douagi et al., 2000).
The epithelial cells within the thymic primordium tinue to proliferate strongly after E12.5, at least partly in response to factors supplied by the mesenchymal capsule
con-Concomitantly, TEC differentiation commences, with the
fi rst evidence of differentiation into cortical and medullary
cell types appearing by E12.5 (Bennett et al., 2002)
Trang 31De-velopment of the two compartments then proceeds in a
lymphocyte-independent manner until E15.5 (Klug et al.,
2002; Jenkinson et al., 2005) The expression of MHC II
and MHC I on the surface of thymic epithelial cells is fi rst
detected at E13.5 and ∼E16, respectively (Jenkinson et al.,
1981; Van Vliet et al., 1984), and is followed by the
appear-ance of CD4+ and CD8+ SP thymocytes at E15.5 and E17.5
(Jenkinson et al., 1981; Van Vliet et al., 1984) Although a
functional thymus is present in neonates, the full
organiza-tion of the stroma is not achieved until two to three weeks
postnatally in the mouse
Origin of Thymic Epithelial Cells
The precise embryonic origins of the thymic
epithe-lium were until recently a matter of long-standing
contro-versy; confl icting hypotheses suggested that the epithelium
had a dual endodermal/ectodermal origin (Cordier and
Heremans, 1975; Cordier and Haumont, 1980) or derived
solely from the pharyngeal endoderm (Le Douarin and
Jotereau, 1975; Manley and Blackburn, 2003; Blackburn and
Manley, 2004; Gordon et al., 2004) However, recent work
from our laboratories has provided defi nitive evidence for a
single endodermal origin in mice (Gordon et al., 2004)
through histological, fate, and potency analysis of the
pha-ryngeal region These data demonstrated that although the
3PP endoderm and third pharyngeal cleft ectoderm make
contact at E10.5, as proposed in the dual-origin hypothesis,
the germ layers subsequently separate, with apoptosis
oc-curring in the contact region Lineage tracing of pharyngeal
surface ectoderm of E10.5 mouse embryos also failed to fi nd
evidence for an ectodermal contribution to the thymic
pri-mordium, and, fi nally, transplantation of pharyngeal
endo-derm isolated from E8.5–E9.0 embryos (i.e., prior to initiation
of overt thymus organogenesis) indicated that the grafted
endoderm was suffi cient for complete thymus
organogene-sis, similar to previous results obtained using chick-quail
chimeras (Le Douarin and Jotereau, 1975) Thus, pharyngeal
endoderm alone is suffi cient for the generation of both
corti-cal and medullary thymic epithelial compartments
Thymic Epithelial Progenitor Cells (TEPC)
The phenotype of TEPC has been of considerable
interest Evidence suggestive of a progenitor/stem cell
activity was initially provided by analysis of a subset of
human thymic epithelial tumours that were found to contain
cells that could generate both cortical and medullary
sub-populations This suggested that the tumourigenic targets
were epithelial progenitor/stem cells (Schluep et al., 1988)
In addition, ontogenic studies suggested that the early
thymus primordium in both mouse and human might be
characterized by coexpression of markers that later
segre-gated to either the cortical or medullary epithelium (Lampert
and Ritter, 1988) However, the fi rst genetic indication of a
TEPC phenotype was provided by a study addressing the
nature of the defect in nude mice, which fail to develop a
functional thymus due to a single base deletion in the
tran-scription factor Foxn1 (Blackburn et al., 1996) Here, sis of allophenic nude-wild-type aggregation chimeras demonstrated that cells homozygous for the nude mutation
analy-were unable to contribute to the major thymic epithelial
subsets, establishing that the nude gene product (Foxn1) is
required cell-autonomously for the development and/or
maintenance of all mature TEC However, a few
nude-derived cells were present in the thymi of adult chimeras, and phenotypic analysis indicated that these cells expressed
determinants reactive to mAbs MTS20 (Godfrey et al., 1990)
and MTS24 but did not express markers associated with mature TEC, including MHC Class II Based on these fi nd-
ings, we suggested that in the absence of Foxn1, TEC lineage
cells undergo maturational arrest and persist as MTS20+24+
progenitors (Blackburn et al., 1996) This hypothesis was
recently confi rmed by an elegant study that demonstrated that functional thymus tissue containing organized cortical and medullary regions is generated on reactivation of a con-ditional null allele of Foxn1 in the postnatal thymus (Bleul
et al., 2006) Since clonal reactivation of Foxn1 was achieved
in this study, it demonstrates unequivocally that in the absence of Foxn1, some persisting TECs have bipotent pro-genitor activity
Further data regarding the phenotype of thymic lial progenitors came from analysis of mice, with a second-ary block in thymus development resulting from a primary
epithe-T-cell differentiation defect The thymi of postnatal CD3e26tg
mice, in which thymocyte development is blocked at the CD44+CD25− TN1 stage (Hollander et al., 1995), contain
principally epithelial cells that coexpress K5 and K8 (Klug
et al., 1998) — which, in the normal postnatal thymus, are
predominantly restricted to the medulla and cortex tively and are coexpressed by only a small population of cells at the corticomedullary junction In this study, Klug and colleagues (1998) demonstrated that transplantation of
respec-CD3e26tg thymi into Ragl −/− mice, which sustain a later block
in T-cell differentiation, resulted in the development of K5−K8+ cells, suggesting that the K5+K8+ cells are progenitors
of cTEC
More recently, two studies have addressed the typic and functional properties of MTS20+24+ cells within the fetal mouse thymus directly Ontogenic analysis has demonstrated that the proportion of MTS20+24+ epithelial cells was highest in the early thymus primordium, decreas-
pheno-ing to less than 1% in the postnatal thymus (Bennett et al.,
2002), consistent with the expression profi le expected of markers of fetal tissue progenitor cells Phenotypic analysis
of the MTS20+24+ and MTS20−24− populations of the E12.5 thymus indicated that all cells in the MTS20+24+ population coexpressed K5 and K8, while none expressed TEC differen-
tiation markers, including MHC II (Bennett et al., 2002)
Importantly, the functional capacity of isolated MTS20+24+cells and MTS20−24− cells was then determined via ectopic transplantation This analysis demonstrated that
I V T H Y M U S O R G A N O G E N E S I S • 651
Trang 32the MTS20+24+ population was suffi cient for establishment
of a functional thymus containing both cortical and
medullary TEC populations, while, in this assay, the
MTS20−24− population fulfi lled none of these functions
(Bennett et al., 2002; Gill et al., 2002) The studies described
earlier clearly demonstrated that progenitor activity resided
in the MTS20+24+ population However, although these data
strongly suggested the existence of common thymic
epi-thelial progenitor cells, this was not addressed at the clonal
level in either study
With regard to this issue, a recent study indicates that
during initial organogenesis a bipotent progenitor exists
that can form both cortical and medullary TECs and
sug-gests that this activity may persist in the postnatal thymus
(Bleul et al., 2006) The approach used was to perform a
lineage trace using hK14CreERt2 transgenic mice crossed onto
a ROSA26/silent eYFP background In this system,
sponta-neous Cre recombinase activity in thymic epithelial cells
activated the expression of eYFP in a small number of TECs
at around 14 days postpartum Analysis of the thymi by fl
uo-rescence microscopy revealed that the majority of eYFP+
cells were present in clusters and were not evenly
distrib-uted throughout the stroma, suggesting that fl uorescent
cells were derived from a single recombination event in cells
with proliferative capacity The location of the cell clusters
was mixed; a small proportion of eYFP+ clusters were
restricted to either the cortex or the medulla, but the
major-ity (76%) appeared to span the corticomedullary junction
These results are consistent with the presence of a bipotent
progenitor that may give rise to intermediate progenitors
committed to either the cortical or medullary lineage
However, a major caveat for this interpretation is that stem
cell activity per se was not assayed in these experiments and
that the data would also be consistent with proliferation of
differentiated epithelial cells In an elegant extension of
these experiments, the same hK14CreERt2 deletor strain was
used to reactivate a conditional null allele of Foxn1 in
post-natal mice (discussed earlier) Here, clonal activation of
Foxn1 resulted in the generation of small regions of thymus
tissue that contained both cortical and medullary TEC,
pro-viding conclusive evidence for the existence of a common
progenitor in initial organogenesis
Human Thymus Development
Early human thymus development closely parallels that
of the mouse Thus, the thymus forms from the third
pha-ryngeal pouch in a common primordium with the
parathy-roid gland The third pharyngeal pouch is evident from early
in week 6 of human fetal development, and initially it
devel-ops as a tubelike lateral expansion from the pharynx, which
makes contact with the ectoderm of the third pharyngeal
cleft (Weller, 1933; Norris, 1938) A single endodermal origin
has not been demonstrated directly for the human thymic
epithelium However, since the thymus has a single
endo-dermal origin in mice and avians (Le Douarin and Jotereau,
1975), it is reasonable to assume that this is also the case in humans Within the human common thymus/parathyroid primordia, the thymus and parathyroid domains are located ventrally and dorsally and are surrounded by condensing neural crest–derived mesenchyme from the onset of devel-opment The thymus component of this primordium begins
to migrate ventrally from week 7 to mid-week 8, forming a highly lobulated, elongated, cordlike structure The upper part of this structure normally disappears at separation of the two organ rudiments, leaving the parathyroid in the approximate location in which it will remain throughout adulthood (Norris, 1938) The bilateral thymic primordia continue to migrate toward the midline, where they eventu-ally meet and attach at the pericardium — the permanent location of the thymus into adulthood — by mid-week 8 (Norris, 1938) As in the mouse, the human early thymus primordium appears to contain undifferentiated epithelial
cells (Bennett et al., 2002), which express some markers that
are later restricted to either cortical or medullary ments (Lampert and Ritter, 1988; A Farley and CCB, unpub-lished data) Nascent medullary development is evident from week 8, and by week 16 distinct cortical and medullary compartments are present Other cell types penetrate the thymus from week 8, including mesenchymal, vascular and lymphoid cells, and mature lymphocytes begin to leave the thymus to seed the peripheral immune tissues between weeks 14 and 16 (van Dyke, 1941; Lobach and Haynes, 1986)
compart-Cervical Thymus in Mouse and Human
The presence of a cervical thymus in humans has been recorded for some time (van Dyke, 1941; Tovi and Mares, 1978; Ashour, 1995), and recent publications indicate that
an ectopic cervical thymus is also a common occurrence in
at least some mouse strains (Dooley et al., 2006; Terszowski
et al., 2006) In terms of size and cellularity, the cervical
thymus is much smaller than the thoracic thymus However, the morphology of the two structures is very similar, with organized cortical and medullary regions and similar expres-
sion patterns of cytokeratin molecules (Dooley et al., 2006;
Terszowski et al., 2006) Furthermore, the cervical thymus
expresses the transcription factors Foxn1 and Aire and can produce functional T-cells that are tolerant to self-antigens
(Dooley et al., 2006; Terszowski et al., 2006).
The origin of the cervical thymus is at present unclear
A plausible hypothesis is that it may arise from remnants of the thymus domain of the 3PP that become detached from the organ during separation of the thymus and parathyroid domains Several alternative explanations exist, and the identifi cation of cervical thymi in mice will allow the embry-onic origins of these structures to be addressed experimen-tally It appears that in mice the cervical thymus may mature
postnatally (Terszowski et al., 2006), while in humans it is
clearly present in the second trimester of fetal development
Although the presence of Foxn1+ epithelial cells has not
Trang 33been reported in the cervical regions of developing mouse
embryos, because it is now clear that cells specifi ed to the
thymic epithelial lineage retain their identity in the absence
of Foxn1 expression (Bleul et al., 2006), Foxn1 may not be an
appropriate lineage marker for tracing the origins of this
tissue
Molecular Regulation of Thymus and
Parathyroid Organogenesis
Although the regulation of thymus organogenesis is
incompletely understood, studies of classical and
geneti-cally engineered mouse mutants have begun to reveal a
network of transcription factors and signaling molecules
that act in the pharyngeal endoderm and surrounding
mesenchyme and mesoderm to regulate thymus and
parathyroid organogenesis The principal components of this network are discussed next and are summarized in Fig 44.2
Molecular Control of Early Organogenesis
The earliest events in thymus organogenesis occur prior
to overt organ development and relate to molecular control
of 3PP formation The T-box transcription factor Tbx1, noic acid (RA) signaling, and fi broblast growth factor 8 (Fgf8) signaling have been implicated as important regulators of this process
reti-Tbx1 was recently identifi ed as the gene responsible for cardiovascular and glandular defects in Df1 mice, which
carry a large deletion of chromosome 16 (Lindsay et al.,
1999) Df1 heterozygotes closely phenocopy a human
FIG 44.2. Molecular regulation of early thymus
organogenesis (A) At approximately E8.0–E8.5
the formation of the third pharyngeal pouch (3PP)
is initiated in the pharyngeal endoderm and is
dependent on the expression of Tbx1 and retinoic
acid (RA) and fi broblast growth factor 8 (Fgf8)
signaling (pink) (B) At E9.5 the 3PP has formed
and is surrounded by mesenchymal cells of
meso-dermal and neural crest cell (NCC) origin
Contin-ued development is dependent on the expression
of the transcription factors Hoxa3, Pax1, Pax9,
Eya1, and Six1 (green) (C) Bone morphogenetic
protein (BMP) (blue) and sonic hedgehog (Shh)
(pink) signaling occur at E10.5 in the 3PP
endo-derm in the ventral and dorsal aspects,
respec-tively These factors may be involved in the
specifi cation of the 3PP into thymus- and
para-thyroid-specifi c domains (D) At E11.5 epithelial
cells in the ventral domain of the 3PP express the
transcription factor Foxn1 (light blue) and will
form the thymic epithelium Epithelial cells in the
dorsal domain express the transcription factor
Gcm2 (purple) and will form the parathyroid gland
The differentiation and maintenance of both of
these cell types are dependent on these factors
I V T H Y M U S O R G A N O G E N E S I S • 653
Trang 34condition known as 22q11.2 deletion syndrome (22q11.2DS,
or DiGeorge Syndrome), in which a deletion in
chromo-some 22 covering an interval of approximately 30 genes
(Scambler, 2000) results in a range of defects including
thymus aplasia or, more frequently, hypoplasia (Paylor et al.,
2001; Taddei, Morishima et al., 2001) Thus the Df1 mouse
represents a useful model for 22q11.2DS and has allowed
the identifi cation of Tbx1 as a critical early regulator of
pha-ryngeal development
During development, Tbx1 is expressed in the
geal endoderm and the core mesenchyme of the
pharyn-geal arches from approximately E7.5 and continues to be
expressed in a variety of structures until E12.5 (Chapman et
al., 1996; Hu et al., 2004; Xu et al., 2004) Tbx1 mutants have
severe defects in the pharyngeal region, including abnormal
patterning of the fi rst pharyngeal arch; hypoplasia of the
second arch; and absence of the third, fourth and sixth
arches and pouches (Jerome and Papaioannou, 2001) As a
result of this, Tbx1−/− mutants lack both thymus and
para-thyroid and display a spectrum of cardiovascular
abnor-malities and craniofacial defects (Jerome and Papaioannou,
2001) The phenotype of Tbx1−/− animals suggests an
impor-tant role for this gene in the segmentation of the pharyngeal
region Supporting evidence for this hypothesis was
pro-vided by an elegant study addressing the temporal
require-ment for Tbx1 in the developrequire-ment of the pharyngeal region
Deletion of Tbx1 at E8.5, during the formation of the 3PP,
resulted in complete absence of thymus and parathyroid,
and complementary fate-mapping experiments
demonstrated that cells that express Tbx1 at E8.5 contribute signifi
-cantly to the thymic primordium (Xu et al., 2005) However,
although deletion of Tbx1 at E9.5/E10.5 (after initial
forma-tion of the 3PP) caused morphological defects in the thymus,
these were not as severe as the aplasia seen after deletion at
E8.5, and fate mapping of cells expressing Tbx1 cells at E9.5/
E10.5 revealed only a small contribution to the thymus (Xu
et al., 2005) Taken together, these data suggest that Tbx1
is required for establishment of the 3PP but that it is
not directly required for subsequent thymus development
Thus, Tbx1 may infl uence later stages of thymus
organo-genesis in a non-cell-autonomous manner Furthermore,
although Tbx1 is haplo-insuffi cient with respect to thymus
development (Lindsay et al., 2001), the basis of this insuffi
-ciency remains to be determined and may result either from
secondary effects resulting from mild defects in pouch
for-mation or from dosage effects related to factor provision by
non-NCC mesenchymal cells
A role for RA in 3PP formation was suggested by
experi-ments in which RA antagonist was administered to
whole-embryo cultures Here, blockade of RA signaling at E8.0
resulted in the absence of the growth factors Fgf8 and Fgf3
in the 3PP endoderm and impaired NCC migration to the
third and fourth pharyngeal arches (Wendling et al., 2000)
Expression of the transcription factor Pax9 (see later) was
also absent in the third pouch, but was expanded in the
second pouch endoderm These data suggest that RA ing is required for the specifi cation of the third pouch endo-derm, which confers subsequent competence to support
signal-NCC migration In vivo evidence for a role for RA signaling
was subsequently provided by the fi nding that fetal mice lacking RA receptors α and β display thymus agenesis and
ectopia (Ghyselinck et al., 1997).
There is considerable evidence that Fgf8 is required during the early stages of thymus and parathyroid develop-ment Fgf8 is expressed in the early gut endoderm and in the endoderm and ectoderm of the pharyngeal pouches and clefts Mice carrying hypomorphic alleles of Fgf8 show defects in thymus development ranging from hypoplasia to
complete aplasia (Abu-Issa et al., 2002; D U Frank et al.,
2002): The initial impairment in thymus and parathyroid organogenesis is likely to occur at an early stage in develop-ment in these mice, because the third and fourth pharyn-geal arches and pouches are usually hypoplastic/aplastic, in addition to other abnormalities in the pharyngeal region
(Abu-Issa et al., 2002; D U Frank et al., 2002).
In terms of the cell types affected by impaired Fgf8 naling, similarities between the phenotype of Fgf8 hypo-morphs and the spectrum of abnormalities found during experimental NCC ablation (Bockman and Kirby, 1984;
sig-Conway et al., 1997) suggest that the glandular defects may
result from defective NCC migration/differentiation or vival In support of this, NCC of Fgf8 hypomorphs show
sur-increased levels of apoptosis (Abu-Issa et al., 2002; D U
Frank et al., 2002) and reduced expression of Fgf10 (Frank
et al., 2002), a factor that may mediate proliferation of the
pharyngeal endoderm (D U Frank et al., 2002) There is
also a mild reduction in the expression of genes associated
with differentiated NCC (Abu-Issa et al., 2002), suggesting
that the maintenance of NCC is perturbed Taken together, these data implicate Fgf8 in maintaining a competent NCC population that can contribute to thymus organogenesis
However, Fgf8 may also act specifi cally on the 3PP derm, since ablation of Fgf8 in the endoderm and ectoderm
endo-or ectoderm alone results in different phenotypes; ablation
in the ectoderm alone causes vascular and craniofacial
defects seen in Fgf8 hypomorphs (Macatee et al., 2003),
whereas when Fgf8 is also deleted in the endoderm, glandular defects are evident, including thymus hypo-
plasia and ectopia (Macatee et al., 2003) Since the NCC
defects were the same in both cases, Fgf8 may directly infl uence development of the endoderm, although it remains possible that the differences observed result from
a dosage effect
Transcription Factors and Regulation of 3PP Development
After the initial onset of 3PP formation, continued development is dependent on several transcription factors, most notably Hoxa3, Pax1, Pax 9, Eya1, Six1, and Six4 All of these transcription factors are expressed in the 3PP endo-derm from approximately E9.5 to E10.5 and, with the excep-
Trang 35tion of Pax1 and Pax9, are also expressed in associated NCC
and the ectoderm
Absence of functional Hoxa3 or Eya1 results in the
failure to initiate overt thymus and parathyroid
organogen-esis once the 3PP has formed, revealing the essential roles
of these factors (Manley and Capecchi, 1995, 1998; Xu et al.,
2002; Zou et al., 2006) Tbx1 and Fgf8 are both
down-regulated in the 3PP of Eya1−/− mice at E9.5 (Zou et al., 2006),
indicating that Eya1 plays a role in the regulation of each of
these factors Lack of Six1, Six1 and 4, Pax1, or Pax9 causes
much less severe phenotypes The common primordium
begins to develop in Six1−/− mice, and patterning into thymus
and parathyroid domains (see the next section) is initiated
However, subsequent apoptosis of endodermally derived
cells in the common primordium leads to complete
disap-pearance of the organ rudiment by E12.5 (Zou et al., 2006)
A similar phenotype is evident in Six1−/−;Six4−/− embryos,
though the size of the primordium is further diminished in
the double mutants, indicating synergy between these gene
products (Zou et al., 2006) Loss of function mutations in
Pax9 results in failure of the primordia to migrate to the
mediastinum and in severe hypoplasia from E14.5
How-ever, the thymic lobes are vascularized and contain
lym-phocytes (Hetzer-Egger et al., 2002) Pax1−/− mutants show
relatively mild thymus hypoplasia and aberrant TEC
differ-entiation (Wallin et al., 1996; Su and Manley, 2000; Su et al.,
2001) Since Pax1 and 9 are highly homologous, these
phe-notypes may refl ect functional redundancy, as
demon-strated in other tissues (Peters and Balling, 1999)
Expression of Hoxa3, Eya1 and Six1 in multiple germ
layers complicates interpretation of the respective null
phenotypes for these genes However, Hoxa3 appears to
regulate Pax1 and Pax9 either directly or indirectly, since
Pax1 and Pax9 expression is initiated normally in Hoxa3−/−
mutants but fails to be maintained at wild-type levels
beyond E10.5 (Manley and Capecchi, 1995) Furthermore,
Hoxa3+/−;Pax1−/− compound mutants show delayed
separa-tion of the thymus/parathyroid primordium from the
pharynx, resulting in thymic ectopia and a more severe
hypoplasia than that seen in Pax1−/− single mutants (Su
et al., 2001).
It has been suggested that Eya1 and Six1 act downstream
of the Hox/Pax genes, for Eya1−/− embryos show normal
expression of Hoxa3, Pax1, and Pax9 but reduced expression
of Six1 in the endoderm of the third and fourth pouches
and ectoderm of the second, third, and fourth pharyngeal
arches (Xu et al., 2002) However, while it is likely that Six1
acts downstream of Eya1, recent evidence indicates that
Eya1 and Six1 do not act downstream of the Pax genes This
has been shown by analysis of Pax9−/− and Pax1−/−Pax9−/−
mutants, which show normal expression of Eya1 and Six1
in the 3PP, and of Eya1−/−;Six1−/− double mutants, which
lack expression of Pax1 in the E10.5 3PP (Zou et al., 2006)
Interestingly, Pax9 expression is unaffected in Eya1/Six1
double mutants (Zou et al., 2006) However, it remains
possible that Eya1 and Six1 may be regulated by Hoxa3 pendently of Pax1 and 9 function
inde-The data just reviewed indicate the power of pound-mutant analysis for unraveling genetic interactions However, further work is required to elucidate exactly how these and other transcription regulators cooperate in the development of the 3PP endoderm and to determine the precise role of each of these factors at the cellular level
com-Specifi cation of the Thymus and Parathyroid
Prior to the overt formation of the thymus and roid, the 3PP is specifi ed into organ-specifi c domains At E9.5, epithelial cells within the anterior dorsal aspect initi-ate expression of Gcm2, a transcription factor required for
parathy-the development of parathy-the parathyroid (Gordon et al., 2001)
Current evidence suggests that Gcm2 acts downstream of Eya1 and Hoxa3, because Gcm2 is down-regulated in Hoxa3−/−, Eya1−/− and Hoxa3+/−;Pax1−/− compound mutant
mice (Su et al., 2001; Xu et al., 2002; Blackburn and Manley,
2004) The fi rst thymus specifi c transcription factor, Foxn1,
is expressed at functionally relevant levels in the ventral domain of the 3PP from approximately E11.25, although
low levels can be detected by PCR from E10.5 (Gordon et al., 2001; Balciunaite et al., 2002) Foxn1, a forkhead class tran-
scription factor, is required for TEC differentiation and hair development, and is discussed in more detail in the next section Upstream regulation of Foxn1 is currently not understood, and analysis of mutants in potential upstream regulators have not been informative in this regard Hoxa3−/−and Eya1−/− embryos do not express Foxn1, but this is due to the block in primordium formation prior to the onset of Foxn1 expression Although Foxn1 expression is unaltered
in Pax9−/− and Hoxa3+/−;Pax1−/− mutants (Hetzer-Egger et al.,
2002; Su and Manley, 2000), the possibility remains of dancy between Pax1 and Pax9 Although Six1−/− mutants display reduced Foxn1 expression, the 3PP exhibits increased
redun-cell death in the absence of Six1 (Zou et al., 2006), and
there-fore the reduced expression of Foxn1 may refl ect poor vival of Foxn1+ cells rather than a direct interaction between Foxn1 and Six1 Further studies are required to determine whether a regulatory relationship exists between these genes and Foxn1
sur-The expression patterns of Foxn1 and Gcm2 clearly defi ne the thymus and parathyroid domains of the 3PP However, these factors do not appear to be responsible for specifi cation of their respective organs, indicating that specifi cation must be mediated by an upstream factor or factors With respect to Foxn1, this model is supported by the fi nding that epithelial cells in the ventral domain of the 3PP express the thymus specifi c cytokine IL-7, a factor required for thymocyte differentiation throughout develop-
ment, at E11.5 (von Freeden-Jeffry et al., 1995; Zamisch et
al., 2005), making IL-7 one of the earliest currently
identi-fi ed markers of thymus identity In the thymus primordium, IL-7 is regulated independently of Foxn1, because Foxn1−/−
I V T H Y M U S O R G A N O G E N E S I S • 655
Trang 36embryos show normal IL-7 expression (Zamisch et al., 2005),
indicating that thymus identity is specifi ed in the absence
of Foxn1 Transplantation experiments also support this
model, because E9.0 pharyngeal endoderm, which has not
yet formed the 3PP, gives rise to a functional thymus when
grafted ectopically (Gordon et al., 2004), indicating that at
this developmental stage some cells are already specifi ed to
the thymic epithelial lineage Although Rhox4 has
previ-ously been identifi ed as an early marker of thymus identity
in the 3PP, the human orthologues of this gene are not
expressed in human thymus organaogenesis, making it
unlikely that it plays a critical role in lineage specifi cation
(Morris et al., 2006).
Similarly, our recent studies conclude that Gcm2 is not
required for specifi cation of the parathyroid, for other
para-thyroid-specifi c markers, including CCL21 and CaSR, are
initiated but not maintained in Gcm2−/− mice (Z Liu, S Yu,
and N R M., unpublished) Thus, Gcm2 may play an
analo-gous role in parathyroid development to that of Foxn1 in the
thymus
How, then, are the thymus and parathyroid domains
within the 3PP established? Evidence suggests that
oppos-ing gradients of bone morphogenetic proteins (BMP) and
sonic hedgehog (Shh) may play an important role in this
process During thymus development, BMP4 expression is
fi rst detected at E9.5, when it is expressed by a small number
of mesenchymal cells in the third pharyngeal arch (Patel
et al., 2006) By E10.5, the BMP4 expression domain has
expanded to include the ventral 3PP endoderm and the
adjacent mesenchyme but remains absent from the dorsal
3PP This expression pattern is maintained at E11.5, and by
E12.5 BMP4 is expressed throughout the thymic
primor-dium and the surrounding mesenchymal capsule (Patel
et al., 2006) The expression pattern of BMP4 suggests that
it may be responsible for the initiation of Foxn1 expression,
because BMP4 is restricted to the ventral domain of the
3PP immediately prior to the onset of Foxn1 expression
Furthermore, Noggin expression is restricted to the dorsal
anterior region of the 3PP at E10.5 and E11.5 and there is
some in vitro evidence that BMP4 can directly regulate
Foxn1 expression (Tsai et al., 2003; A Farley and C C B.,
unpublished data)
In vivo evidence of a role for BMPs in thymus
organo-genesis has been provided by a transgenic approach in
which the BMP inhibitor Noggin is driven by the Foxn1
pro-moter, thus impairing BMP signaling in the thymic stroma
(Bleul and Boehm, 2005) These mice have hypoplastic and
cystic thymi that fail to migrate to their normal position
above the heart It is highly likely that this is due to a direct
effect on the thymic stroma, because impaired development
is evident prior to the immigration of lymphocytes, which
are able to differentiate into T-cells Furthermore, mediators
of BMP signaling, such as Msx1 and phosphorylated
Smad proteins, are down-regulated in both the epithelium
and surrounding mesenchyme, suggesting that impaired
communication between these cell types is the mechanism responsible for the phenotype observed Interestingly, Foxn1 expression was not overtly affected in this transgenic model, arguing against a role for BMP in the regulation of this tran-scription factor However, the inhibition of BMP signaling is driven by the Foxn1 promoter, and thus it may occur after the point at which it is required for initiation of Foxn1 expression In addition, since BMP2 and BMP7 are also expressed in the 3PP and common primordium (C C B and
N R M., unpublished data), it is highly likely that dancy operates between different BMP family members It may thus be more pertinent to view BMPs in terms of the signaling they mediate during organogenesis rather than which specifi c member of the BMP family is involved
redun-During development of the 3PP, expression of the secreted glycoprotein Shh is restricted to cells of the pouch opening at E10.5 and E11.5, although its receptor, Patched1,
is expressed by cells in close proximity to this region Scott and Manley, 2005) Analysis of Shh−/− embryos revealed that both the BMP4 and Foxn1 expression domains are expanded in the 3PP, while the corresponding Gcm2+ para-thyroid domain is lost in these mutants (Moore-Scott and Manley, 2005) Thus, the role of Shh in thymus organogen-esis may be to oppose the action of BMP4 to allow the speci-
(Moore-fi cation and development of the parathyroid
Wnt glycoproteins may also be important in regulating Foxn1 expression Wnts are expressed by the thymic stroma and lymphoid cells, although Wnt receptors are expressed
exclusively by TECs (Balciunaite et al., 2002) The earliest
reported expression of Wnt family members during thymus development is at E10.5, immediately prior to strong Foxn1 expression, when the epithelium of the 3PP and adjacent
cells express Wnt4 (Balciunaite et al., 2002) Given this
expression pattern and the fi nding that TEC lines that
over-express Wnt4 display elevated levels of Foxn1 (Balciunaite et
al., 2002), it is possible that Wnt4 cooperates with BMP4 to
regulate Foxn1 Wnt 1, Wnt 4, and Wnt1, 4-null mice all exhibit hypoplastic thymi characterized by reduced T-cell numbers but normal thymocyte developmental progres-sion However, because no histological analysis of the thymi
in these mutants has been presented, it is not possible to evaluate whether the primary effect is on the thymic epithe-
lium or on thymocytes (Mulroy et al., 2002; Staal and Clevers,
2005) As with BMP family members, functional redundancy
is again a possibility, because other Wnt family members are also expressed in the 3PP and surrounding mesenchyme
(Mulroy et al., 2002; Balciunaite et al., 2002; C C B and
N R M., unpublished data)
Foxn1 and the Regulation of TEC Differentiation
As discussed earlier, the development of functionally mature TECs from the 3PP endoderm is cell autonomously
dependent on Foxn1 (Blackburn et al., 1996) Adult nude
mice, which lack functional Foxn1, retain a cystic, phoid thymus consisting predominantly of apparently
Trang 37alym-immature epithelial cells (Cordier, 1974; Cordier and
Haumont, 1980; Gordon et al., 2001) These and other data
(see earlier) suggest that lack of Foxn1 results in
develop-mental arrest of thymic-epithelial-lineage cells at the
founder/progenitor-cell stage of development Thus,
whereas TEPCs form independent of Foxn1, their
differen-tiation into mature TEC subtypes depends on it In the
thymus, Foxn1 is expressed by all thymic epithelial cells
throughout development and is maintained postnatally
(Nehls et al., 1996) It is also expressed in the hair follicles
and epidermis, where it is required for normal development
of the skin (Flanagan, 1966)
The precise role of Foxn1 has not been completely
elu-cidated in either the thymic or the cutaneous epithelial
lineages However, considerable evidence suggests that it
regulates the balance between epithelial proliferation and
differentiation Keratinocytes derived from Foxn1−/− mice
have reduced proliferative capacity in vitro and prematurely
express markers associated with terminal differentiation
(Brissette et al., 1996) Furthermore, when Foxn1 is
over-expressed, markers associated with earlier stages of
differ-entiation are up-regulated and later markers are absent
(Brissette et al., 1996) More recently it was shown that
primary human keratinocytes can be induced to initiate
ter-minal differentiation by transient expression of Foxn1 but
that completion of the differentiation program is dependent
on the levels of activated Akt, which may in turn be
regu-lated by Foxn1 (Janes et al., 2004) Thus, in the epidermis
Foxn1 may function to ensure that terminal differentiation
proceeds in a temporally regulated manner Whether this
is the case in the thymus is unclear The early thymic
rudiment in nude mice also shows reduced proliferation
(Itoi et al., 2001) However, the epithelial cells resemble
immature progenitors and do not express markers of
terminal differentiation
There is some evidence to suggest that in the thymus,
Foxn1 is required for lymphoepithelial cross-talk
Compara-tive gene expression analysis between nude and wild-type
thymus suggests that PD1 Ligand is a target of Foxn1,
because it is down-regulated in nude embryos (Bleul and
Boehm, 2001) The receptor for PD1 ligand is expressed by
thymocytes and has been implicated in thymocyte survival,
proliferation and positive selection (Nishimura et al., 1996,
2000), lending weight to the hypothesis that Foxn1 may
regulate genes required for lymphoepithelial cross-talk
A separate publication suggested a role for Foxn1 in
cross-talk by analyzing Foxn1∆/∆ mice, which express a splice
variant lacking exon 3 of the N-terminal domain (Su et al.,
2003) Skin and hair development is normal in these mice,
but the differentiation of TECs is suspended at a time
equi-valent to E13.5 Unlike the nude thymus, the thymic stroma
can attract lymphocytes and mediate T-cell differentiation,
although there are postnatal abnormalities in thymocyte
maturation and a severe reduction in thymocyte numbers
The thymus-specifi c defect in Foxn1∆/∆ mice resembles to
some degree the phenotype seen in hCD3ε26 mice, which have secondary blocks in TEC maturation due to suspended
thymocyte development (Klug et al., 1998, 2002) Based on
this, we suggested that the N-terminal domain is required for lymphoepithelial cross-talk and as such has a thymus-specifi c function However, the impairment of TEC differen-tiation in Foxn1∆/∆ mice is more profound than in hCD3ε26 mutants and occurs prior to the stage when TEC develop-ment is dependant on lymphoepithelial cross-talk It is therefore unclear if Foxn1 directly regulates genes involved
in cross-talk or if its primary role is to regulate TEC entiation, which has downstream effects on lymphoepithe-lial communication
differ-Molecular Regulation of TEC Proliferation
Following the formation of the thymic primordium and the commitment of the epithelial cells to the TEC lineage, the thymus undergoes a period of expansion involving both the proliferation of stromal cells and an increase in thymocyte numbers With regard to TECs, the growth factors
Fgf7 and Fgf10 have been shown to be involved In vitro
experiments have demonstrated that both Fgf7 and Fgf10, which are expressed by the perithymic mesenchyme, can
stimulate the proliferation of fetal TECs (Suniara et al., 2000)
Furthermore, mice lacking Fgfr2IIIb, the receptor for Fgf7 and Fgf10, have severely hypoplastic thymi, although they can support T-cell maturation, and Fgf10−/− mutants also
develop hypoplastic thymi (Revest et al., 2001).
Noncanonical NF-kB Signaling Regulates mTEC Development
The development of mTECs depends on activation of the NF-kB signaling pathway, specifi cally the noncanonical pathway that culminates in RelB activation Medullary TEC development is severely compromised in RelB-defi cient
mice (Burkly et al., 1995; Weih et al., 1995) In the
nonca-nonical RelB activation pathway, ligand engagement of receptors in the TNFR family, such as lymphotoxinβ recep-tor (LTβR), activates NF-kB-inducing kinase (NIK), which phosphorylates homodimers of the downstream kinase, Ikkα Activated Ikkα in turn phosphorylates the C-terminal region of NF-kB2 (p100), leading to ubiquitin-dependent degradation and release of the N-terminal polypeptide, p52 The formation of RelB/p52 heterodimers permits shuttling
of RelB from the cytoplasm into the nucleus, where it
func-tions as a transcriptional regulator (Bonizzi et al., 2004) A
range of medullary defects occurs in mice that are defi cient
in various components upstream of RelB in the alternative NF-kB activation pathway Targeted disruption of the LTβR gene results in disorganized medullary regions that contain
reduced numbers of both major mTECs subsets (Boehm et
al., 2003) Mice with a naturally occurring mutation in NIK
(Barcena et al., 1994) and Ikkα knockout mice have severe defects in medullary formation, including impairment of mTECs development and reduced expression of AIRE and
I V T H Y M U S O R G A N O G E N E S I S • 657
Trang 38tissue-restricted antigens (Kajiura et al., 2004; Kinoshita
et al., 2006; E Richie, unpublished observations) A similar
medullary phenotype is found in TRAF6 knockout
mice (Akiyama et al., 2005) Interestingly, each of these
mutant strains develops autoimmune manifestations,
indicating a breakdown in the establishment of central
tolerance
Role of Medullary Thymic Epithelial Cells in
Establishing Central Tolerance
Medullary TECs have the unique ability to express genes
encoding a wide array of antigens that initially were thought
to be expressed only in peripheral tissues (Derbinski et al.,
2001) AIRE is expressed by mTECs and plays a prominent
role in regulating tissue-restricted antigen expression In
humans, homozygous AIRE mutations result in a severe,
multiorgan autoimmune disease termed autoimmune
poly-glandular syndrome type 1 (Villasenor et al., 2005) AIRE
knockout mice develop a similar autoimmune phenotype
due to defective clonal deletion, resulting in persistence
of self-reactive thymocytes (Anderson et al., 2002; Liston
et al., 2003) Although AIRE is clearly important for
regula-tion of tissue-restricted antigen expression in mTECs, other
as-yet-undefi ned factors are involved since certain
tissue-restricted antigens are expressed even in the absence of
AIRE (Derbinski et al., 2005) Medullary TECs also affect
central tolerance by supplying self-peptides to medullary
dendritic cells (DCs) in a process termed cross-presentation
(Gallegos and Bevan, 2004) Medullary TECs are also required
for the generation of CD4+CD25+ T regulatory (Treg) cells
and natural killer T-cells, both of which actively repress
self-reactive T-cells (Kronenberg and Rudensky, 2005; Kim et al.,
2006) Given that AIRE and tissue-restricted antigens are
highly expressed by mTECs, it is likely that the autoimmune
phenotype in mice defi cient in various components of the
RelB activation pathway is a result of failed mTEC ment Taken together it is now clear that mTECs contribute directly and indirectly to establishing central tolerance and averting the development of autoimmunity
develop-V SUMMARY
Thymus organogenesis is a complex process in which a three-dimensional organ forms from the endoderm of the 3PP The thymic epithelium, a critical regulator of thymo-poiesis, comprises many subtypes of TEC, all of which arise from a common progenitor Recent studies have begun to clarify the lineage relationships between these cell types and to identify the molecular factors that govern thymus organogenesis However, several signifi cant questions remain unresolved:
• What mechanisms regulate formation of the 3PP from the endoderm?
• How does the network of transcription factors and naling molecules that are expressed in the endoderm and surrounding mesenchyme/mesoderm control sub-sequent 3PP development?
sig-• What factor or factors specify thymus and parathyroid lineages within the 3PP?
• What are the factors responsible for the maintenance, proliferation and differentiation of TEPCs?
Additionally, important questions remain regarding static maintenance of the mature thymus, including eluci-dation of whether the postnatal organ is maintained by a stem cell mechanism, and the cellular and molecular mech-anisms that operate to induce thymic involution Resolution
homeo-of these issues will permit the design homeo-of rational strategies for therapeutic reconstitution of the adaptive immune system The development of such strategies should signifi -cantly impact the health of the aging population and other immunocompromised individuals
VI ACKNOWLEDGMENTS
We wish to thank Lucy Morris (Carnegie Institution of
Washington, D.C, and Howard Hughes Medical Institute,
Baltimore), for critical reading of the manuscript, and our
funding bodies, Leukaemia Research (CCB, CSN), the EU (CCB, CSN), and the NIH (NRM, ER), for support
VII REFERENCES
Abu-Issa, R., Smyth, G., et al (2002) Fgf8 is required for pharyngeal
arch and cardiovascular development in the mouse Development
129(19), 4613–4625.
Akiyama, T., Maeda, S., et al (2005) Dependence of self-tolerance on
TRAF6-directed development of thymic stroma Science 308(5719),
248–251.
Anderson, G., Owen, J J., et al (1994) Thymic epithelial cells provide
unique signals for positive selection of CD4+CD8+ thymocytes in vitro
J Exp Med 179(6), 2027–2031.
Anderson, M S., Venanzi, E S., et al (2002) Projection of an
immuno-logical self-shadow within the thymus by the aire protein Science
298(5597), 1395–1401.
Ashour, M (1995) Prevalence of ectopic thymic tissue in myasthenia
gravis and its clinical signifi cance J Thorac Cardiovasc Surg 109(4),
632–635.
Balciunaite, G., Keller, M P., et al (2002) Wnt glycoproteins regulate the expression of FoxN1, the gene defective in nude mice Nat Immunol
3(11), 1102–1108.
Baldwin, T A., Sandau, M M., et al (2005) The timing of TCR alpha
expression critically infl uences T-cell development and selection
J Exp Med 202(1), 111–121.
Barcena, A., Galy, A H., et al (1994) Lymphoid and myeloid
differentia-tion of fetal liver CD34+-lineage cells in human thymic organ culture
J Exp Med 180(1), 123–132.
Trang 39Bennett, A R., Farley, A., et al (2002) Identifi cation and
characteriza-tion of thymic epithelial progenitor cells Immunity 16(6), 803–814.
Blackburn, C C., and Manley, N R (2004) Developing a new paradigm
for thymus organogenesis Nat Rev Immunol 4(4), 278–289.
Blackburn, C C., Augustine, C L., et al (1996) The nu gene acts
cell-autonomously and is required for differentiation of thymic epithelial
progenitors Proc Natl Acad Sci U.S.A 93(12), 5742–5746.
Bleul, C C., and Boehm, T (2001) Laser capture microdissection-based
expression profi ling identifi es PD1-ligand as a target of the nude locus
gene product Eur J Immunol 31(8), 2497–2503.
Bleul, C C., and Boehm, T (2005) BMP signaling is required for normal
thymus development J Immunol 175(8), 5213–5221.
Bleul, C C., Corbeaux, T., et al (2006) Formation of a functional thymus
initiated by a postnatal epithelial progenitor cell Nature 441(7096),
992–996.
Bockman, D E., and Kirby, M L (1984) Dependence of thymus
devel-opment on derivatives of the neural crest Science 223(4635), 498–500.
Boehm, T., Scheu, S., et al (2003) Thymic medullary epithelial cell
dif-ferentiation, thymocyte emigration, and the control of autoimmunity
require lympho-epithelial cross-talk via LTbetaR J Exp Med 198(5),
757–769.
Bofi ll, M., Janossy, G., et al (1985) Microenvironments in the normal
thymus and the thymus in myasthenia gravis Am J Pathol 119(3),
462–473.
Bonizzi, G., Bebien, M., et al (2004) Activation of IKKalpha target genes
depends on recognition of specifi c kappaB binding sites by RelB:p52
dimers Embo J 23(21), 4202–4210.
Boyd, R L., Wilson, T J., et al (1992) Phenotypic characterization of
chicken thymic stromal elements Dev Immunol 2(1), 51–66.
Boyd, R L., Tucek, C L., et al (1993) The thymic microenvironment
Immunol Today 14(9), 445–459.
Brekelmans, P., and van Ewijk, W (1990) Phenotypic characterization
of murine thymic microenvironments Semin Immunol 2(1), 13–24.
Brissette, J L., Li, J., et al (1996) The product of the mouse nude locus,
Whn, regulates the balance between epithelial cell growth and
differ-entiation Genes Dev 10(17), 2212–2221.
Burkly, L., Hession, C., et al (1995) Expression of relB is required for
the development of thymic medulla and dendritic cells Nature
373(6514), 531–536.
Chapman, D L., Garvey, N., et al (1996) Expression of the T-box family
genes, Tbx1–Tbx5, during early mouse development Dev Dyn 206(4),
379–390.
Clark, R A., Yamanaka, K., et al (2005) Human skin cells support
thymus-independent T cell development J Clin Invest 115(11),
3239–3249.
Conway, S J., Henderson, D J., et al (1997) Pax3 is required for cardiac
neural crest migration in the mouse: evidence from the splotch (Sp2H)
mutant Development 124(2), 505–514.
Cordier, A C (1974) Ultrastructure of the thymus in “Nude” mice
J Ultrastruct Res 47(20), 26–40.
Cordier, A C., and Haumont, S M (1980) Development of thymus,
parathyroids, and ultimo-branchial bodies in NMRI and nude mice
Am J Anat 157(3), 227–263.
Cordier, A C., and Heremans, J F (1975) Nude mouse embryo:
ectodermal nature of the primordial thymic defect Scand J Immunol
4(2), 193–196.
Cumano, A., Dieterlen-Lievre, F., et al (1996) Lymphoid potential,
probed before circulation in mouse, is restricted to caudal
intraembry-onic splanchnopleura Cell 86(6), 907–916.
De Smedt, M., Reynvoet, K., et al (2002) Active form of Notch imposes
T cell fate in human progenitor cells J Immunol 169(6), 3021–3029.
Derbinski, J., Schulte, A., et al (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self Nat
Immunol 2(11), 1032–1039.
Derbinski, J., Gabler, J., et al (2005) Promiscuous gene expression in
thymic epithelial cells is regulated at multiple levels J Exp Med 202(1),
Douagi, I., Andre, I., et al (2000) Characterization of T cell precursor
activity in the murine fetal thymus: evidence for an input of T cell
pre-cursors between days 12 and 14 of gestation Eur J Immunol 30(8),
2201–2210.
Farr, A G., and Nakane, P K (1983) Cells bearing Ia antigens in the
murine thymus An ultrastructural study Am J Pathol 111(1), 88–97.
Flanagan, S P (1966) “Nude,” a new hairless gene with pleiotropic
effects in the mouse Genet Res 8(3), 295–309.
Frank, D U., Fotheringham, L K., et al (2002) An Fgf8 mouse mutant
phenocopies human 22q11 deletion syndrome Development 129(19),
4591–4603.
Frank, J., Pignata, C., et al (1999) Exposing the human nude phenotype
[letter] Nature 398(6727), 473–474.
Gallegos, A M., and Bevan, M J (2004) Driven to autoimmunity: the
nod mouse Cell 117(2), 149–151.
Ghyselinck, N B., Dupe, V., et al (1997) Role of the retinoic acid
recep-tor beta (RARbeta) during mouse development Int J Dev Biol 41(3),
425–447.
Gill, J., Malin, M., et al (2002) Generation of a complete thymic
micro-environment by MTS24(+) thymic epithelial cells Nat Immunol 3(7), 635–642.
Godfrey, D I., Izon, D J., et al (1990) The phenotypic heterogeneity of
mouse thymic stromal cells Immunology 70(1), 66–74.
Gordon, J., Bennett, A R., et al (2001) Gcm2 and Foxn1 mark early
parathyroid- and thymus-specifi c domains in the developing third
pharyngeal pouch Mech Dev 103(1–2), 141–143.
Gordon, J., Wilson, V A., et al (2004) Functional evidence for a single
endodermal origin for the thymic epithelium Nat Immunol 5(5),
546–553.
Hare, K J., Jenkinson, E J., et al (1999) In vitro models of T cell
development Semin Immunol 11(1), 3–12.
Hetzer-Egger, C., Schorpp, M., et al (2002) Thymopoiesis requires
Pax9 function in thymic epithelial cells Eur J Immunol 32(4), 1175–
1181.
Hollander, G A., Wang, B., et al (1995) Developmental control point in
the induction of thymic cortex regulated by a subpopulation of
prothy-mocytes Nature 373, 350–353.
Hu, T., Yamagishi, H., et al (2004) Tbx1 regulates fi broblast growth
factors in the anterior heart fi eld through a reinforcing autoregulatory
loop involving forkhead transcription factors Development 131(21),
5491–5502.
V I I R E F E R E N C E S • 659
Trang 40Itoi, M., Kawamoto, H., et al (2001) Two distinct steps of immigration
of haematopoietic progenitors into the early thymus anlage Int
Immunol 13, 1203–1211.
Janes, S M., Ofstad, T A., et al (2004) Transient activation of FOXN1
in keratinocytes induces a transcriptional program that promotes
terminal differentiation: contrasting roles of FOXN1 and Akt J Cell Sci
117(Pt 18), 4157–4168.
Jenkinson, E J., and Owen, J J (1990) T-cell differentiation in thymus
organ cultures Semin Immunol 2(1), 51–58.
Jenkinson, E J., Van Ewijk, W., et al (1981) Major histocompati bility
complex antigen expression on the epithelium of the developing
thymus in normal and nude mice J Exp Med 153(2), 280–292.
Jenkinson, E J., Anderson, G., et al (1992) Studies on T cell maturation
on defi ned thymic stromal cell populations in vitro J Exp Med 176(3),
845–853.
Jenkinson, W E., Rossi, S W., et al (2005) Development of functional
thymic epithelial cells occurs independently of lymphostromal
interac-tions Mech Dev 122(12), 1294–1299.
Jerome, L A., and Papaioannou, V E (2001) DiGeorge syndrome
phe-notype in mice mutant for the T-box gene, Tbx1 Nat Genet 27(3),
286–291.
Jiang, X., Rowitch, D H., et al (2000) Fate of the mammalian cardiac
neural crest Development 127(8), 1607–1616.
Jotereau, F., Heuze, F., et al (1987) Cell kinetics in the fetal mouse
thymus: precursor cell input, proliferation, and emigration J Immunol
138(4), 1026–1030.
Kajiura, F., Sun, S., et al (2004) NF-kappa B-inducing kinase establishes
self-tolerance in a thymic stroma-dependent manner J Immunol
172(4), 2067–2075.
Kim, H J., Hwang, S J., et al (2006) NKT cells play critical roles in the
induction of oral tolerance by inducing regulatory T cells producing
IL-10 and transforming growth factor beta, and by clonally deleting
antigen-specifi c T cells Immunology 118(1), 101–111.
Kinoshita, D., Hirota, F., et al (2006) Essential role of IkappaB kinase
alpha in thymic organogenesis required for the establishment of
self-tolerance J Immunol 176(7), 3995–4002.
Klug, D B., Carter, C., et al (1998) Interdependence of cortical thymic
epithelial cell differentiation and T-lineage commitment Proc Natl
Acad Sci U.S.A 95(20), 11822–11827.
Klug, D B., Carter, C., et al (2002) Cutting edge:
thymocyte-indepen-dent and thymocyte-depenthymocyte-indepen-dent phases of epithelial patterning in the
fetal thymus J Immunol 169(6), 2842–2845.
Kronenberg, M., and Rudensky, A (2005) Regulation of immunity by
self-reactive T cells Nature 435(7042), 598–604.
Kurobe, H., Liu, C., et al (2006) CCR7-dependent cortex-to-medulla
migration of positively selected thymocytes is essential for establishing
central tolerance Immunity 24(2), 165–177.
La Motte-Mohs, R N., Herer, E., et al (2005) Induction of T-cell
devel-opment from human cord blood hematopoietic stem cells by Delta-like
1 in vitro Blood 105(4), 1431–1439.
Lampert, I A., and Ritter, M A (1988) The origin of the diverse
epithe-lial cells of the thymus: is there a common stem cell? In “Thymus
Update” (M D Kendall and M A Ritter, eds.), pp 5–25 Harwood
Aca-demic, London.
Le Douarin, N M., and Jotereau, F V (1975) Tracing of cells of the avian
thymus through embryonic life in interspecifi c chimeras J Exp Med
142(1), 17–40.
Le Lievre, C S., and Le Douarin, N M (1975) Mesenchymal derivatives
of the neural crest: analysis of chimaeric quail and chick embryos
J Embryol Exp Morphol 34(1), 125–154.
Lehar, S M., and Bevan, M J (2002) T cell development in culture
Immunity 17(6), 689–692.
Lind, E F., Prockop, S E., et al (2001) Mapping precursor movement
through the postnatal thymus reveals specifi c microenvironments
sup-porting defi ned stages of early lymphoid development J Exp Med
194(2), 127–134.
Lindsay, E A., Botta, A., et al (1999) Congenital heart disease in mice
defi cient for the DiGeorge syndrome region Nature 401(6751),
379–383.
Lindsay, E A., Vitelli, F., et al (2001) Tbx1 haploinsuffi cieny in the DiGeorge syndrome region causes aortic arch defects in mice Nature
410(6824), 97–101.
Liston, A., Lesage, S., et al (2003) Aire regulates negative selection of
organ-specifi c T cells Nat Immunol 4(4), 350–354.
Lobach, D F., and Haynes, B F (1986) Ontogeny of the human thumus
during fetal development J Clin Immunol 7, 81–97.
Macatee, T L., Hammond, B P., et al (2003) Ablation of specifi c
expres-sion domains reveals discrete functions of ectoderm- and derived FGF8 during cardiovascular and pharyngeal development
endoderm-Development 130(25), 6361–6374.
Manley, N R., and Blackburn, C C (2003) A developmental look at thymus organogenesis: where do the non-hematopoietic cells in the
thymus come from? Curr Opin Immunol 15(2), 225–232.
Manley, N R., and Capecchi, M R (1995) The role of Hoxa-3 in mouse
thymus and thyroid development Development 121(7), 1989–2003.
Manley, N R., and Capecchi, M R (1998) Hox group 3 paralogs late the development and migration of the thymus, thyroid, and para-
regu-thyroid glands Dev Biol 195(1), 1–15.
Moore-Scott, B A., and Manley, N R (2005) Differential expression of Sonic hedgehog along the anterior–posterior axis regulates patterning
of pharyngeal pouch endoderm and pharyngeal endoderm-derived
organs Dev Biol 278(2), 323–335.
Morris, L., Gordon, J., et al (2006) Identifi cation of a tandem
duplicated array in the Rhox locus on mouse chromosome X
Mammalian Genome, in press.
Mulroy, T., McMahon, J A., et al (2002) Wnt-1 and Wnt-4 regulate
thymic cellularity Eur J Immunol 32(4), 967–971.
Nehls, M., Kyewski, B., et al (1996) Two genetically separable steps in
the differentiation of thymic epithelium Science 272(5263), 886–889.
Nishimura, H., Agata, Y., et al (1996) Developmentally regulated
expression of the PD-1 protein on the surface of double-negative (CD4–
CD8–) thymocytes Int Immunol 8(5), 773–780.
Nishimura, H., Honjo, T., et al (2000) Facilitation of beta selection and
modifi cation of positive selection in the thymus of PD-1-defi cient mice
Owen, J J., and Ritter, M A (1969) Tissue interaction in the
develop-ment of thymus lymphocytes J Exp Med 129(2), 431–442.
Patel, S R., Gordon, J., et al (2006) Bmp4 and Noggin expression during
early thymus and parathyroid organogenesis Gene Expr Patterns 6(8),
794–799.