+ All Categories
Home > Documents > Isolated Islets in Diabetes Research

Isolated Islets in Diabetes Research

Date post: 04-Jun-2018
Category:
Upload: mohammad-sutami
View: 220 times
Download: 0 times
Share this document with a friend
16
Isolated islets in diabetes research R. Bhonde, R.C. Shukla, M. Kanitkar, R. Shukla, M. Banerjee & S. Datar Tissue Engineering & Banking Laboratory, National Centre for Cell Science, Pune, India Received November 8, 2006 This review highlights some recent developments and diversified applications of islets in diabetes research as they are rapidly emerging as a model system in biomedical and biotechnological research. Isolated islets have formed an effective in vitro model in antidiabetic drug development programme, screening of potential hypoglycaemic agents and for investigating their mechanisms of action. Yet another application of isolated islets could be to understand the mechanisms of  cell death in vitro and to identify the sites of intervention for possible cytoprotection. Advances in immunoisolation and immunomodulation protocols have made xeno-transplantation feasible without immunosuppression thus increasing the availability of islets. Research in the areas of pancreatic and non pancreatic stem cells has given new hope to diabetic subjects to renew their islet cell mass for the possible cure of diabetes. Investigations of the factors leading to differentiation of pancreatic stem/progenitor cells would be of interest as they are likely to induce pancreatic regeneration in diabetics. Similarly search for the beta cell protective agents has a great future in preservation of residual beta cell mass left after diabetogenic insults. We have detailed various applications of islets in diabetes research in context of their current status, progress and future challenges and long term prospects for a cure. Key words Cytoprotection - hypoglycaemics - islets - regeneration - transplantation Islets of Langerhans are organelles present within the pancreas and are mainly responsible for the production of insulin, glucagon, somatostatin and pancreatic polypeptide upon stimulation. The primary focus of islet research is, however, the cure and/or better management of diabetes mellitus which results from a loss of insulin secretion from beta cells present within the islets of Langerhans. This review seeks to take a bird’s eye view of the contribution of islets as a model system in diabetes research beyond transplantation. Since their discovery in 1869 1 , islets have been viewed as a possible in vitro system for a syndrome that cannot be mimicked very effectively using cell lines. They are miniature organ systems, retaining their architecture, differentiated state and ability of insulin secretion upon stimulation, independent of nervous control. Isolation of islets 2,3 has promoted studies related to understanding the pathophysiology of type I and II diabetes, transplantation, screening of hypoglycaemic drugs and probing into diabetes causing mechanisms, to device effective means of prevention. 42 5 Indian J Med Res 125, March 2007, pp 425-440
Transcript

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 1/16

Isolated islets in diabetes research

R. Bhonde, R.C. Shukla, M. Kanitkar, R. Shukla, M. Banerjee & S. Datar

Tissue Engineering & Banking Laboratory, National Centre for Cell Science, Pune, India

Received November 8, 2006

This review highlights some recent developments and diversified applications of islets in diabetes

research as they are rapidly emerging as a model system in biomedical and biotechnological research.

Isolated islets have formed an effective in vitro model in antidiabetic drug development programme,

screening of potential hypoglycaemic agents and for investigating their mechanisms of action. Yet

another application of isolated islets could be to understand the mechanisms of  cell death in vitro

and to identify the sites of intervention for possible cytoprotection. Advances in immunoisolation

and immunomodulation protocols have made xeno-transplantation feasible without

immunosuppression thus increasing the availability of islets. Research in the areas of pancreatic

and non pancreatic stem cells has given new hope to diabetic subjects to renew their islet cell massfor the possible cure of diabetes. Investigations of the factors leading to differentiation of pancreatic

stem/progenitor cells would be of interest as they are likely to induce pancreatic regeneration in

diabetics. Similarly search for the beta cell protective agents has a great future in preservation of 

residual beta cell mass left after diabetogenic insults. We have detailed various applications of 

islets in diabetes research in context of their current status, progress and future challenges and

long term prospects for a cure.

Key words Cytoprotection - hypoglycaemics - islets - regeneration - transplantation

Islets of Langerhans are organelles present within

the pancreas and are mainly responsible for the

production of insulin, glucagon, somatostatin andpancreatic polypeptide upon stimulation. The primary

focus of islet research is, however, the cure and/or

better management of diabetes mellitus which results

from a loss of insulin secretion from beta cells present

within the islets of Langerhans. This review seeks to

take a bird’s eye view of the contribution of islets as a

model system in diabetes research beyond

transplantation.

Since their discovery in 18691, islets have been

viewed as a possible in vitro  system for a syndrome

that cannot be mimicked very effectively using celllines. They are miniature organ systems, retaining their

architecture, differentiated state and ability of insulin

secretion upon stimulation, independent of nervous

control. Isolation of islets2,3 has promoted studies related

to understanding the pathophysiology of type I and II

diabetes, transplantation, screening of hypoglycaemic

drugs and probing into diabetes causing mechanisms,

to device effective means of prevention.

425

Indian J Med Res 125, March 2007, pp 425-440

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 2/16

Islets in pharmacological research

 Insulin secretion enhancers: Isolated islets, in vitro,

respond to glucose stimulation and hence haveimmensely contributed to the study of various

pharmacological aspects and for screening of 

promising antidiabetic agents. A number of 

hypoglycaemic drugs act as insulin secretagogues,

in corroboration with this, we have reported that islets

can serve as an in vitro model for antidiabetic drug

screening4. Our model offers several advantages as

it is simple and economical in terms of reduction in

the number of animals used as well as the amount of 

drug required for testing. A number of plants, in

traditional medicine, are claimed to have anti-

diabetic properties and isolated islets have been used

extensively for checking their properties and

plausible modes of action. For example, extract of 

Tinospora crispa5, extract of Gymnema sylvestre6,

bittergourd fruit juice7, leaf extract of Urtica diocia8,

aqueous extract of Scoparia dulcis9  and aqueous

extract of Teucrium polium10  have been shown to

exhibit insulin secretagogue activity. Apart fromplant/natural extracts, several synthetic drugs have

been tested for their insulin secreatagogue property

and for determination of their mechanism of action,

using isolated pancreatic islets in vitro.

It is understood that glucose stimulates insulin

secretion in the pancreatic   cell by means of a

synergistic interaction between at least two signaling

pathways. In the K (ATP) channel-dependent

pathway, glucose stimulation increases the entry of 

extrinsic Ca2+  through voltage-gated channels by

closure of the K (ATP) channels and depolarization

of the beta cell membrane. The resulting increase in

intracellular Ca2+  stimulates insulin exocytosis.

While in the GTP-dependant pathway, intracellular

Fig. 1. Signaling cascade of glucose stimulated insulin secretion. The figure summarizes the signaling pathway triggered by glucose

in beta cells for insulin exocytosis. Agents in blue represent secretagogues while agents in red represent insulin secretion inhibitors.

Gq protein: member of G family of proteins, IP3: Inositol triphosphate.

426 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 3/16

Ca2+  is elevated by GTP-dependent proteins and

augments the Ca2+-stimulated release (Fig. 1).

Secretagogues and insulin secretion inhibitors act at

intermediate steps of these signaling pathways andinfluence the process of insulin exocytosis. Several

researchers have investigated this intricate mode of 

known secretagogue action using isolated islets as

an in vitro  model. To quote a few; imidazoline

antagonists of alpha 2-adrenoreceptors increase

insulin release in vitro  by inhibiting ATP-sensitive

K+ channels in pancreatic   cells11. Giannaccini et 

al12  have evaluated the properties of sulphonylurea

receptors (SUR) of human islets of Langerhans. They

studied the binding affinity of various oral

hypoglycaemic agents to the receptor and also tested

insulinotropic action of the drugs on intact human

islets. This binding potency order was parallel with

the insulinotropic potency of the evaluated

compounds12 . Masuda et al13   have shown an

insulinotropic effect of Triglitazone (CS-045) and

have shown its mode of action to be distinct from

glibenclamide (a sulphonylurea drug). A-4166, a

derivative of D-phenylalanine, evokes a rapid and

short-lived hypoglycaemic action in vivo. It has been

shown to act via the tolbutamide binding sites 14.Louchami et al15  showed S21403, a meglitinide

analogue to be a novel insulinotropic tool in the

treatment of type 2 diabetes, as it affected cationic

fluxes and the drugs secretary responses displayed

favourable time course of prompt, and not unduly

prolonged, activation of   cells. Mears et al16

demonstrated that tetracaine (an anaesthetic)

stimulates insulin secretion by release of intracellular

calcium and for the first time elucidated the role of 

intracellular calcium stores in stimulus-secretion

coupling in the pancreatic   cells. JTT-608, is a

nonsulphonylurea oral hypoglycaemic agent which

stimulates insulin release at elevated but not low

glucose concentrations by evoking PKA-mediated

Ca2+ influx17.

 Insulin secretion inhibitors: Besides insulinotropic

studies, several investigators have used isolated islets

to study drugs that inhibit insulin secretion and their

probable mode of action. Several drugs have

inhibitory effect on insulin secretion by  cells and

this is an important aspect, which needs to be

considered while using the said drug. Cyclosporine,

a widely used immunosuppressant, induces inhibitionof insulin release in isolated rat islets18,19. Paty et al20

demonstrated the inhibitory effects of several

immunosuppressive drugs on insulin secretion. Thus

low dose immunosuppressive drug protocols should

be used in clinical islet transplantation and patients

using this drug have to be carefully monitored for

signs of deficient insulin secretion. Tacrolimus is

known to cause post-transplant diabetes mellitus.

Using isolated rat pancreatic islets, Uchizono et al21

studied its mechanism of action and found that it

interferes with the process of insulin exocytosis and

that protein kinase C (PKC)-mediated (Ca2+

dependent and independent) and Ca2+-independent

GTP signaling pathways may be involved. L-

asparaginase inhibits glucose-induced insulin

secretion in a dose-dependent manner by decreasing

total cAMP in isolated rat islets22. Metz et al23 have

used selective inhibitors of GTP synthesis and proved

that they impede exocytotic insulin release from

intact rat islets. The study provides first direct

evidence that GTP is required for insulin release23.Hydrochlorothiazide is also known to inhibit insulin

release. This inhibition is not mediated by reduced

chloride fluxes but rather by inhibition of calcium

uptake24.

Many antidiabetic drugs act on peripheral tissues

and have no direct effect on pancreatic islets. One

such drug is metformin, which affects glucose and

free fatty acid metabolism in peripheral insulin target

tissues. However, Patane et al25   exposed rat

pancreatic islets to high glucose and free fatty acid

(FFA) levels in vitro   mimicking the in vivo

environment in presence and absence of metformin.

They found that metformin can restore a normal

secretary pattern in islets whose secretary function

has been impaired by chronic exposure to elevated

FFA or glucose levels. Recently, Marchetti et al26

cultured islets isolated from type 2 diabetes subjects

in presence of metformin and found that several

functional and survival defects of T2D islets were

BHONDE et al: ISLETS IN DIABETES RESEARCH 427

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 4/16

ameliorated by metformin. Thus in diabetic patients,

metformin (in addition to its peripheral effects) may

have a direct beneficial effect on the  cell secretory

function. On the contrary, Leclerc et al27 have shownthe ability of metformin to activate AMP-activated

protein kinase in human islets and inhibit insulin

secretion. This inhibitory effect needs to be

considered with respect to the use of this drug for

the treatment of type II diabetes.

We examined the effect of commonly used

antibiotics such as gentamycin, penicillin,

streptomycin, tetracycline, neomycin, erythromycin

and chloramphenicol on isolated islets viability,

functionality and induction of oxidative stress if any.

Our results revealed the innocuous nature of the

antibiotics used at pharmacological concentrations,

suggesting their safety whenever prescribed for

combating infections and also during islet isolation

procedures28.

Cytoprotection of islets

Another primary area of research greatly

facilitated by usage of islets as a model system is inunraveling the mysteries of beta cell death and ways

of preventing the same.

   cell death: The precise mechanisms of  cell death

in vivo, leading to diabetes, remain unclear. However,

extensive studies, using islets as a model system show

that there are many molecules including Fas Ligand

(FasL) and cytokines such as interleukin-1 (IL-1),

tumour necrosis factor alpha (TNF) and interferon

gamma (IFN  ) that cause release of other cytokine

mediators that have potential to damage   cells in

vitro  and in vivo29.   cell death appears to be

ultimately caused by receptor mediated mechanisms

and/or by secretion of cytotoxic molecules like

granzymes and perforin. In addition, toxic molecules

such as reactive oxygen species (ROS: superoxide

radicals, hydroxyl radicals, and nitric oxide) play a

significant role in islet cell death by inducing DNA

damage. DNA damage, in  cells, leads to poly (ADP

ribose) polymerase (PARP) activation which

increases NAD consumption, depletion of which

compromises ATP production in cells30. It is apparent

that a number of different mechanisms of cell death

are operative in destruction of islets (Fig. 2).

 Inhibi tion of ox idants : Normally, cells counter

oxidative stress by expression of ROS scavenging

enzymes like catalase (CAT), glutathione peroxidase

(Gpx) and superoxide dismutase (SOD).   cells,

however, have extraordinarily low levels of ROS

scavenging enzymes31 . The correction of this

deficiency, in vitro, by overexpression of cellular

enzymes like SOD may lead to protection of  cells

against oxidative stress induced   cell damage/ 

death32. Studies, wherein mitochondrial form of Mn-

SOD was overexpressed in cells are shown to have

protected isolated islets against oxidative damage in

vitro33. It has also been shown that adenoviral

overexpression of glutamyl cyesteine ligase catalytic

subunit, a primary regulator of de novo synthesis of 

glutathione (GSH) in mammalian cells and central

to the antioxidant capacity of the cell, protects

pancreatic islets against oxidative stress, in vitro34.

 In vitro stress in islets is often produced by using

cell specific toxins like streptozotocin (STZ) andalloxan. STZ induces islet necrosis by employing

effector molecules like nitric oxide (NO) and ROS.

Pro-inflammatory cytokines also employ NO as an

effector molecule for necrosis and / or apoptosis

induction 35 . Hence a plausible means for

cytoprotection of islets could be scavenging of NO

or inhibition of iNOS (inducibe nitric oxide synthase)

which synthesizes NO. iNOS inhibitors can be used

for cytoprotection of islets in vitro as inhibition of 

iNOS would inhibit formation of NO, preventing islet

cell death indirectly. It has been reported that a

combination of an iNOS inhibitor and a free radical

scavenger, guanidinoethyldisulphide restored IL-1

induced suppression of islet insulin secretion in

vitro36. An imidazole compound called Efaroxan has

also been shown to impart complete protection

against IL-1 induced toxicity37. It has recently been

shown that silymarin, a polyphenolic flavonoid that

has a strong antioxidant activity, prevented IL-

1+IFN-  -induced NO production and -cell

428 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 5/16

dysfunction in human islets. These cytoprotective

effects of silymarin appeared to be mediated through

the suppression of c-Jun NH2-terminal kinase and

Janus kinase/signal transducer and activator of transcription pathways38.  In vitro treatment of islets

with exogenous antioxidants is another viable option.

There are many known antioxidants like vitamin C,

vitamin E39, Scoparia dulcis a traditional antidiabetic

plant40  that are known to protect islets against

oxidative stress induced dysfunction and death.

Studies with islets have shown that the experimental

drug, bis-o- hydroxycinnamoyl methane, an analogue

of naturally occurring bis- demethoxycurcumin,

enhances the antioxidant defense against

ROS, thus protecting  cells from death41. It has been

demonstrated that polyenoylphosphatidylcholine

(PPC), a phosphatidylcholine rich phospholipid

extracted from soybean, protects  cells against STZ

induced toxicity and also plays an important part in

maintaining their insulin synthesis and secretion for

normal glucose homeostasis42.

PARP and NF   B inhibition : Poly (ADP ribose)

polymerase (PARP) is a major effector molecule in

the oxidative stress induced or cytokine induced cell

death pathway. It is known that STZ injections cause

extensive necrosis in islets of Parp + / + mice while

the extent of necrosis was markedly lower in Parp- / -

islets43. It has also been shown that inhibition of 

Parp-1 by synthetic inhibitors like 3-aminobenzamide

resulted in protection against necrosis. A potent

inhibitor of Parp, 5-iodo-6amino-1, 2 benzopyrone

(INH2BP), was found to protect rat islets and beta

cell line RIN-5F from cytokine induced damage44.

Fig. 2. Mechanisms of islet cell death. Flow chart depicts apoptotic and necrotic beta cell death cascades along with possible modes

of intervention. Causes/agents of beta cell death are indicated in red while agents/strategies for prevention of beta cell damage are

indicated in blue. Red arrows stand for possible sites of intervention. IL-11: Interleukin 11, IKK: Inhibitor of kappa kinase, iNOS:

inducible nitric oxide synthase, NO, Nitric oxide, STZ: Streptozotocin, ROS: Reactive oxygen species, PARP: Poly (ADP-Ribose)

Polymerase, CAT: Catalase: SOD, superoxide dismutase; GSH: glutathione peroxidase, NF-kB: Nuclear Factor kappa B. This figure

has been compiled and constructed by authors by referring to data cited in references 34-38.

BHONDE et al: ISLETS IN DIABETES RESEARCH 429

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 6/16

NF-B activation is an important event in

inflammation, cellular death signaling and is often

activated by oxidative stress45. One of the key steps

in activation of the NF-B pathway is thestimulation of the I-B kinases46. Inhibition of NF-

B activation could be an effective means of 

controlling islet cell death in vitro. Studies have

shown that IL-11, a regulatory cytokine, has been

effective in inhibiting NF-B activation in islets

leading to prevention of multiple low doses STZ

(MLD- STZ) induced diabetes in vivo47. Rehman

et al48  have demonstrated that adenoviral gene

transfer of the NF-B inhibitor I-B to isolated

human islets resulted in protection from IL-1

mediated dysfunction and apoptosis. Mouse islets

when transduced in s i tu by infusion of the

transduction peptide prior to isolation lead to 40

per cent of peptide transduction of  cells. Delivery

of the IKK inhibitor transduction fusion peptide

(PTD-5-NBD) in situ to mouse islets resulted in

improved is let function and viabil i ty af ter

isolation48. Other studies involving rhIL-1  have

suggested a cytoprotective role of the recombinant

cytokine against alloxan induced toxicity in

diabetic rat islets49.

Apart from exogenously induced islet damage,

islets also suffer from damage due to endogenous

hyperglycaemia in vivo. It is known that high glucose

concentration causes apoptosis in cultured human

pancreatic islets of Langerhans. Data suggest that in

human islets, high glucose modulates the balance of 

pro- and anti-apoptotic Bcl proteins towards   cell

apoptosis50. Hyperglycaemia also causes production

of IL-1  by islet   cells leading to cytotoxicity in

human pancreatic islets51. Chronic exposure to free

fatty acids alone or with hyperglycaemic conditions

lead to pancreatic cell death possibly employing

oxidative stress as the mechanism for cell

destruction52,53. Hence, blockage of multiple

pathways, rather than a single pathway, leading to

cell death may be necessary to fully protect  cells

from destruction in vitro54. It is relatively easy to

study mechanisms of   cell death, in its intricate

detail, along with modes of preventing the same and

its effect on diabetogenesis employing isolated islets

rather than animal experimentation.

Isolated islets for transplantation

Apart from studying strategies for prevention or

abrogation of  cell death, isolated islets are widely

used for transplantation. Extensive studies have been

conducted wherein islets were transplanted into a

hyperglycaemic host and then checked for reduction

in hyperglycaemia. Till date it remains the most

successful means of achieving normoglycaemia in

humans55. Allo- or xeno- tranplantation of whole

pancreas is possible56, but it requires major surgery,

hence transplantation of islets57,58 or insulin producing

beta cells59,60 would be a more viable option.

Any successful transplantation depends on three

things, viz:

(i) Primary non function: Primary nonfunctioning

(PNF) of islets accounts for the bulk of graft losses.

Macrophages, the main effector cells in PNF,

release proinflammatory cytokines like IL-1 and

TNF-, which in turn recruit free radicals to mediatea nonspecific inflammatory response61. Lee et al62

have shown that the blockade of monocyte chemo-

attractant protein-1 (MCP-1) binding to CCR2, in

conjunction with sub therapeutic

immunosuppression, leads to islet allograft survival.

Hence, interruption of the leukocyte recruitment

through chemokine receptor targeting may be of 

therapeutic benefit. Transfection of cytoprotective

genes to isolated pancreatic islets may contribute

to enhanced survival in transplant settings, e.g., the

overexpression of erythropoietin gene protects islets

from destruction and does not compromise islet

functionality63. It has been shown by Riachy et al64

using cell lines and human islets, that 1, 25-(OH)

2D3, the active metabolite of vitamin D3, was able

to induce and maintain high levels of A20, an anti-

apoptotic protein known to block NF-B activation,

thus promoting islet cell survival by modulating the

effects of inflammatory cytokines, which contribute

to  cell demise64. Disruption of islet extracellular

430 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 7/16

matrice during pancreatic digestion leads to

induction of apoptotic pathways, thus increasing

cumulative PNF. Targeting the apoptotic pathway

by adenovirus-mediated gene transfer of the anti-apoptotic Bcl-2 gene exerts a major cytoprotective

effect on isolated islets. This was experimentally

proved with isolated macaque pancreatic islets. Bcl-

2 transfection ex vivo  protected these islets from

apoptosis65.

(ii) Abrogation of immune rejection of graft and 

recurrence of autoimmunity: The next hurdle, graft

rejection, can be dealt by the administration of 

immunosuppressive drugs in the host, and/or

immunoisolation of the graft, or immunomodulation

of the graft or host or both 66 . Immuno-

compromisation enables allogenic and xenogenic

islet transplantation in preclinical, non human

primate models 67. Isolated islets are known to

reverse diabetes in immunocompromised nude mice

rendered diabetic by STZ. Although attractive, an

immunosuppression regime leaves the host

susceptible to other infections68, and these drugs

have adverse effects on insulin secretion by

  cells 22,23,58 . A better alternative toimmunocompromization is to make the host tolerant

to the graft. Many different strategies have been

developed to achieve transplantation tolerance. In

one approach used by Oluwole et al 69   the

intravenous administration of genetically

engineered host dendritic cells (DCs) expressing

allo-MHC peptides, along with transient ALS

immunosuppression, resulted in induction of graft

tolerance. Similarly, induction of mixed chimerism

via bone marrow (BM) cells  transplantation from

normal donors into autoimmune non obese diabetic

(NOD) mice has been  shown to reverse insulitis and

prevented the development of diabetes and induces

tolerance to donor islet cells 70-74. This approach

however leaves the host susceptible to the graft

versus host disease (GVHD). Like this, Liang et al75

have described a radiation free regimen for

induction of chimerism, donor-specific tolerance,

reversal of insulitis, and resistance to diabetes

development in NOD mice model.

It is understood that complete T cell activation

requires two signals; T cell receptor (TCR)

interaction with peptide- MHC complex presented

by antigen presenting cells (APCs). This signal mustthen combine with another co-stimulatory signal,

mediated by interaction between distinct cell surface

molecules of APCs and T cells76. In absence of co-

stimulation, T cells undergo anergy and become non-

responsive. The B7/CD28/CTLA4 co-stimulatory

pathway plays a critical role in the regulation of T-

cell activation transplant rejection and autoimmunity.

Adams et al77 have used LEA29Y (BMS-224818), a

mutant of CTLA4-Ig along with repamycin and IL-

2R to effectively prevent the rejection of islet

allograft in a preclinical primate model.

Administration of co-stimulatory blockade (anti-

CD40L) has been   reported to induce mixed

chimerism in NOD mice78,79. Pearson et al80  have

reported that an allelic variant of Idd3 gene is

responsible for prolonged islet allograft survival by

co-stimulatory blockade in NOD mice. MHC

antigens, expressed on APC of donor tissue, stimulate

a higher T cells response as compared to host APCs.

As passenger (donor) APCs are largely responsible

for co-stimulatory activity, graft immunomodulatorystrategies aim at depleting them from the islet grafts81.

These strategies include in vitro  culture of graft at

suboptimal temperature for extended time82,83, UV-

B irradiation84, cryopreservation85,86  mitomycin C

treatment87, ICAM-1 specific monoclonal antibody

treatment88, co-transplantation of islets with sertoli

cells89 etc.

 Xenotransplantation: The existing shortage of donor

islets makes it necessary to pool islets from different

donors or look for alternative islet sources. Foetal,

neonatal and adult porcine islets along with bovine,

murine, canine, avian and piscean islets have been

tested for this purpose and porcine islets have been

found to be an acceptable source for alternative islets.

It was observed that neonatal porcine pancreatic cell

clusters (NPCCs) contain mature endocrine cells and

bring about sustained normalization of blood glucose

levels when transplanted into kidney capsule of 

diabetic nude mice. Rapid return to diabetic state was

BHONDE et al: ISLETS IN DIABETES RESEARCH 431

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 8/16

observed after removal of islet grafts90,91. Recently,

Garkavenko et al92 have reported a follow up study

in human diabetic patients receiving porcine islets

for 9 yr. The finding that none of the patientsdeveloped viral infection hold promises for

xenotransplantation. Similarly, canine, bovine and

porcine islets have been successfully used for

xenotransplantation in a diffusion based bio hybrid

artificial pancreas93,94. Chick embryo pancreatic

transplants have shown reversal in experimental

diabetes of rats without immunosuppression 95.

Transient reversal of experimental diabetes in mice

has also been reported by transplantation

of chicken pancreatic islets96. Xenotransplantation of 

fish islets into the non-cryptorchid testis has also

been carried out. Cryopreservation of principal islets

of teleost fish and their xenotransplantation has also

been studied97,98. Immunoisolation of islets by micro-

encapsulation is of great clinical potential in the

treatment of diabetes with xenotransplantation of 

islets99-102. Lim and Sun, first described the alginate

micro-encapsulation of islets 10 2. Since then

immunoisolation has been regarded as the

technological key to xenotransplantation without

immunosuppression. The encapsulated isletscan be transplanted in, and retrieved from, the

peritoneal cavity with minimal invasive surgery.

Immunoisolation has facilitated the transplantation,

and consequent reversal of hyperglycaemia, from rat

to mice103,104, monkey to rats105, porcine tomurine, dog

to mice106, and even across a large species barrier

i.e., from rabbit to cynomolgus monkey107. An ideal

membrane for immunoisolation should be

biocompatible, non immunogenic, non cytotoxic,

differentially permeable to glucose, insulin, oxygen

and other growth factors required for prolonged

survival of graft and impermeable to

immunoglobulins, immune effector cells and their

recruiting cytokines108. Various natural biopolymers

like and synthetic materials have been used

extensively as immunoisolation material. We have

tested cellulose macrocapsules and molecular

dialysis membrane109,110, polyurethan111, chitosan-

PVP112 and chitosan-alginate113,114 microcapsules for

islet storage and transplantation purposes. Polymeric

biomaterials such as alginate115, agarose116, polyamide

4-6 membranes11 7, poly (ethylene glycol)

diacrylate118, polyvinylchloride acrylic copolymer119,

AN69120, polyurethane-polydimethylsiloxane121, poly(2-hydroxyethylmetacrylate) 122  have also been

proposed as immunobarriers.

Despite these successes the hurdles to be

conquered are monumentous. Theoretically it would

be best if an individual could simply regenerate its

own pancreas/islets.

 cell replication and/or regeneration

Pancreatic islet   cell growth can be mediated

by two separate mechanisms123. Either new islets can

generate from budding of the pancreatic ductal

epithelium 124-128  or from intra islet precursor

cells129,130, i.e., islet-neogenesis and by replication of 

existing islet beta cells123,131 (Fig. 3).

 Is let neogenesis : Neogenesis of islets primarily

occurs during foetal and perinatal stages of 

development132, but has also been observed in the

regenerating adult pancreas133,134. In a population of well differentiated adult pancreatic islet cells, the

number of  cells actually undergoing cell division

is small, measured to be between 0.5 to 2 per cent135.

Although the growth potential of the pancreatic islet

  cells is limited, glucose (nutrients), c-AMP, and

certain polypeptide growth factors have been

reported to exert modest stimulatory effects on  cell

growth and replication136 . Several authors have

reported differential effects of various growth factors

on islet neogenesis phenomenon. Movassat et al137,

have investigated the effects of keratinocytes growth

factor (KGF), in vitro, on  cell differentiation from

undifferentiated pancreatic precursor cells. However

KGF does not help in  cell replication137. Similarly

vascular endothelial growth factor (VEGF- ligand of 

foetal liver kinase-1), has been shown to play a role

in the development of foetal rat islet-like structures

in vitro, possibly by stimulating the maturation of 

endocrine precursor cells in the pancreatic ductal

epithelium138. Epidermal growth factor (EGF), an

432 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 9/16

activator of the MAP kinase pathway, increases the

mass of pancreatic epithelial cells but the absolute

number of developing endocrine cells decreases. On

the other hand, inhibition of MAPK pathway byPD98059 in the precursor cells leads to decreased

proliferation of epithelial cells but endocrine cell

differentiation was activated. Hence, MAPK pathway

determines the final mass of developing endocrine

tissue139.

    cell replication: Sjoholm et al140, reported that

lithium treatment stimulates rat   cell replication

and long term insulin secretion in vitro. The

relationship between  cell replication and insulin

release was further investigated using neonatal rat

pancreatic monolayer cell cultures and the study

demonstrates the importance of glucose utilization

for both of these  cell processes141. In another study

authors have demonstrated that even after complete

destruction of   cells by STZ treatment in vitro,

foetal pancreatic cells retain the ability to regenerate  cells142. The potential for large scale production

of endocrine tissue in vitro  has been indicated,

however, more investigation needs to be carried out

on the various signals and pathways involved in

pancreatic development. An attempt to transduce

NPI (neonatal pancreatic islet) with gene of interest

i.e., PDX-1, allowed researchers to determine the

effects on islet maturation. The authors believed that

these transduced NPIs provide an effective tool to

study islet growth and maturation143. Transfection

of   cells with tyrosine kinase receptors144   and

human islets with chimeric signaling receptors145

leads to ligand dependent cell proliferation. This

Fig. 3.  Islet beta cell birth. Figure depicts different ways of islet beta cell neogenesis. Inset 1 to 4 depicts various stages in islet

generation from pancreatic duct epithelium stem cell monolayer (1), intense zone of activity (2), islet formation (3) and fully formed

mature islet (4). Further inset sequence represents trans-differentiation of islets from non-pancreatic stem cells viz. bone marrow,

acinar cells and hepatic progenitor cells. Insets 5: monolayer, 6: islet-like cell clusters and 7: Budding and maturation of islets.

BHONDE et al: ISLETS IN DIABETES RESEARCH 433

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 10/16

strategy has potential to reduce the quantity of 

human islets required for treatment of patients with

type 1 diabetes.

Conclusion

It is apparent that isolated islets form a handy

model system due to ease of isolation and

maintenance. Being miniature organ systems, they

do not require a nervous control and manipulations

like transfection studies related to signaling

pathways, insulin stimulation and secretion assays

are easy to perform in an in vitro system. Along with

ease of handling, studies based on isolated islets can

be extrapolated and data corroborate with related in

vivo  findings, with high efficiency, thus supporting

the 3R principle of ‘Reduction, Refinement and

Replacement’ of animals in biomedical research.

These factors have led to islets being popularly used

as a compatible model system for diabetes and related

research. All above mentioned approaches are

considered in context of their current status, progress,

future challenges or limitations, and long-term

prospects for a cure. Although definitive success is

still at the horizon, the advances reviewed herepredict the future to be bright.

References

1. Dittrich HM, Hahn von Dorsche H. The anatomical and

histological investigation of the pancreas in the 19th century

and pancreas research from discovery of islets (1869) till

the discovery of pancreas -diabetes 1889.  Anat Anz  1978;

143  : 231-41.

2. Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW.

Automated method for isolation of human pancreatic islets.

 Diabetes 1988; 37   : 413-20.

3. Shewade YM, Umrani M, Bhonde RR. Large-scale isolation

of islets by tissue culture of adult mouse pancreas.

Transplant Proc 1999; 31  : 1721-3.

4. Bhonde RR, Parab PB, Sheorin VS. An in vitro model for

screening oral hypoglycemics.  In vitro Cell Dev Biol Anim

1999; 35  : 366-8.

5. Noor H, Hammonds P, Sutton R, Ashcroft SJ. The

hypoglycaemic and insulinotropic activity of Tinospora

crispa: studies with human and rat islets and HIT-T15 B

cells.  Diabetologia  1989; 32  : 354-9.

6. Persaud SJ, Al-Majed H, Raman A, Jones PM. Gymnema

sylvestre  stimulates insulin release in vitro  by increased

membrane permeability.  J Endocrinol   1999; 163  : 207-12.

7. Sitasawad SJ, Shewade Y, Bhonde R. Role of bittergourd

fruit juice in stz-induced diabetic state in vivo and in vitro.

 J Ethnopharmacol 2000; 73  : 71-9.

8. Farzami B, Ahmadvand D, Vardasbi S, Majin FJ, Khaghani

SH. Induction of insulin secretion by a component of Urtica

dioica  leave extract in perifused Islets of Langerhans and

its in vivo effects in normal and streptozotocin diabetic rats. J Ethnopharmacol  2003; 89  : 47-53.

9. Latha M, Pari L, Sitasawad S, Bhonde R. Insulin-

secretagogue activity and cytoprotective role of the

traditional antidiabetic plant Scoparia dulcis  (Sweet

Broomweed).  Life Sci 2004; 75  : 2003-14.

10. Esmaeili MA, Yazdanparast R. Hypoglycaemic effect of 

Teucrium polium: studies with rat pancreatic islets.

 J Ethnopharmacol  2004; 95  : 27-30.

11. Jonas JC, Plant TD, Henquin JC. Imidazoline antagonists

of alpha 2-adrenoceptors increase insulin release in vitro byinhibiting ATP-sensitive K+ channels in pancreatic beta-

cells.  Br J Pharmacol 1992; 107  : 8-14.

12. Giannaccini G, Lupi R, Trincavelli ML, Navalesi R, Betti

L, Marchetti P, et al. Characterization of sulfonylurea

receptors in isolated human pancreatic islets. J Cell Biochem

1998; 71  : 182-8.

13. Masuda K, Okamoto Y, Tsuura Y, Kato S, Miura T, Tsuda

K, et al. Effects of Troglitazone (CS-045) on insulin

secretion in isolated rat pancreatic islets and HIT cells: an

insulinotropic mechanism distinct from glibenclamide.

 Diabetologia  1995; 38   : 24-30.

14. Tsukuda K, Sakurada M, Niki I, Oka Y, Kikuchi M. Insulin

secretion from isolated rat islets induced by the novel

hypoglycemic agent A-4166, a derivative of D-

phenylalanine.  Horm Metab Res 1998; 30  : 42-9.

15. Louchami K, Jijakli H, Malaisse JW. Effect of the meglitinide

analog S21403 on cationic fluxes and insulin release in

perifused rat pancreatic islets exposed to a high concentration

of D-glucose. Pharmacol Res 1999; 40 : 297-300.

434 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 11/16

16. Mears D, Leighton X, Atwater RE. Tetracaine stimulates

insulin secretion from the pancreatic beta-cell by release of 

intracellular calcium. Cell Calcium 1999; 25 : 59-68.

17. Hashiguchi S, Yada T, Arima T. A new hypoglycemic agent,

JTT-608, evokes protein kinase A-mediated Ca(2+)

signaling in rat islet beta-cells: strict regulation by glucose,

link to insulin release, and cooperation with glucagon-like

peptide-1(7-36)amide and pituitary adenylate cyclase-

activating polypeptide.  J Pharmacol Exp Ther  2001; 296   :

22-30.

18. Nielsen JH, Mandrup-Poulsen T, Nerup J. Direct effects of 

cyclosporin A on human pancreatic beta-cells.  Diabetes

1986; 35  : 1049-52.

19. Robertson RP. Cyclosporin-induced inhibition of insulinsecretion in isolated rat islets and HIT cells. Diabetes 1986;

35  : 1016-9.

20. Paty BW, Harmon JS, Marsh CL, Robertson RP. Inhibitory

effects of immunosuppressive drugs on insulin secretion

from HIT-T15 cells and Wistar rat islets. Transplantation

2002; 73  : 353-7.

21. Uchizono Y, Iwase M, Nakamura U, Sasaki N, Goto D, Iida

M. Tacrolimus impairment of insulin secretion in isolated

rat islets occurs at multiple distal sites in stimulus-secretion

coupling.  Endocrinology  2004; 145  : 2264-72.

22. Pou JM, Cervera T, Perez A, Ortiz MA, Arroyo JA. Effect

of L-asparaginase on insulin secretion from isolated rat islets

of Langerhans.  Horm Res 1991; 35  : 155-60.

23. Metz SA, Rabaglia ME, Pintar TJ. Selective inhibitors of 

GTP synthesis impede exocytotic insulin release from intact

rat islets.  J Biol Chem  1992; 267   : 12517-27.

24. Sandstrom PE. Inhibition by hydrochlorothiazide of insulin

release and calcium influx in mouse pancreatic beta-cells.

 Br J Pharmacol 1993; 110: 1359-62.

25. Patane G, Piro S, Rabuazzo AM, Anello M, Vigneri R,

Purrello F. Metformin restores insulin secretion altered by

chronic exposure to free fatty acids or high glucose: a direct

metformin effect on pancreatic beta-cells.  Diabetes   2000;

49  : 735-40.

26. Marchetti P, Guerra S, Marselli L, Lupi R, Masini M, Pollera

M. Pancreatic islets from type 2 diabetic patients have

functional defects and increased apoptosis that are

ameliorated by metformin.  J Clin Endocrinol Metab  2004;

89  : 5535-41.

27. Leclerc I, Woltersdorf WW, Xavier GS, Rowe RL, Cross

SE, Korbutt GS. Metformin, but not leptin, regulates AMP-

activated protein kinase in pancreatic islets: impact on

glucose-stimulated insulin secretion.  Am J Ph ys iol

 Endocrinol Metab  2004; 286 : E1023-31.

28. Shewade Y, Tirth S, Bhonde RR. Pancreatic islet-cell

viabili ty, functionality and oxidative status remain

unaffected at pharmacological concentrations of commonly

used antibiotics in vitro. J Biosci 2001; 26  : 349-55.

29. Dudeck NL, Thomas HE, Mariana L, Sutherland RM,

Allison J, Estella E, et al. Cytotoxic T-cells from T-cell

receptor transgenic NOD8.3 mice destroy beta-cells via the

perforin and Fas pathways.  Diabetes 2006; 55  : 2412-8.

30. Burkart V, Blaeser K, Kolb H. Potent beta-cell protectionin vitro by an isoquinolinone-derived PARP inhibitor. Horm

 Metab Res 1999; 31  : 641-4.

31. Mandrup-Poulsen T. Beta cell death and protection. Ann NY 

 Acad Sci 2003; 1005 : 32-42.

32. Lortz S, Tiedge M, Nachtwey T, Karlsen AE, Nerup J,

Lenzen S. Protection of insulin-producing RINm5F cells

against cytokine-mediated toxicity through over-expression

of antioxidant enzymes.  Diabetes 2000; 49 : 1123-30.

33. Tran PO, Parker SM, LeRoy E, Franklin CC, Kavanagh TJ,

Zhang T. Adenoviral over-expression of theglutamylcysteine ligase catalytic subunit protects pancreatic

islets against oxidative stress.  J Biol Chem  2004; 279   :

53988-93.

34. Kaneto H, Fujii J, Seo H, Suzuki K, Matsuoka T, Nakamura

M. Apoptotic cell death triggered by Nitric oxide in

pancreatic -cells.  Diabetes 1995; 44 : 733-8.

35. Mabley JG, Southan GJ, Salzman AL, Szabo C. The

combined inducible nitric oxide synthase inhibitor and free

radical scavenger guanidinoethyldisulfide prevents multiple

low-dose streptozotocin-induced diabetes in vivo  and

interleukin-1beta-induced suppression of islet insulin

secretion in vitro. Pancreas 2004; 28  : E39-44.

36. Papaccio G, Graziano A, Valiante S, D’Aquino R, Travali

S, Nicoletti F. Interleukin (IL)-1beta toxicity to islet beta

cells: Efaroxan exerts a complete protection.  J Cell Physiol

2005; 203  : 94-102.

37. Szabo C, Mabely JG, Moeller SM, Shimanovich R, Pacher

P, Virag L, et al. Pathogenetic role of peroxynitrite in the

development of diabetes and diabetic vascular

BHONDE et al: ISLETS IN DIABETES RESEARCH 435

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 12/16

complications: studies with FP15, a novel peroxynitrite

decomposition catalyst.  Mol Med  2002; 8   : 571-80.

38. Matsuda T, Ferreri K, Todorov I, Kuroda Y, Smith CV,

Kandeel F, et al. Silymarin protects pancreatic beta-cells

against cytokine-mediated toxicity: implication of c-Jun

NH2-terminal kinase and janus kinase/signal transducer and

activator of transcription pathways.  Endocrinology  2005;

146   : 175-85.

39. Garg M, Bansal D. Protective antioxidant effects of vitamins

C and E in streptozotocin induced diabetic rats.  Indian J 

 Exp Biol 2000; 38  : 101-4.

40. Latha M, Pari L, Sitasawad S, Bhonde R. Scoparia dulcis, a

traditional antidiabetic plant, protects against streptozotocin

induced oxidative stress and apoptosis in vitro and in vivo. J Biochem Mol Toxicol 2004; 18   : 261-72.

41. Srivivasan A, Menon VP, Periaswamy V, Rajasekaran KN.

Protection of pancreatic beta-cell by the potential antioxidant

bis-o-hydroxycinnamoyl methane, analogue of natural

curcuminoid in experimental diabetes.  J Pharm Pharm Sci

2003; 6   : 327-33.

42. Lee SH, Han YM, Min YM, Park IS., Cytoprotective effects

of polyenoylohosphatidylcholine (PPC) on beta cells during

diabetic induction by streptozotocin. J Histochem Cytochem

2003; 51  : 1005-15.

43. Burkart V, Wand Z, Radons J, Heller B, Herceg Z, Stingl L,

et al. Mice lacking the poly (ADP ribose) polymerase gene

are resistant to pancreatic beta cell destruction and diabetes

development induced by streptozotocin.  Nat Med  1999; 5 :

314-9.

44. Mabely J, Suarez-Pinzon W, Hasko G, Salzmann AL,

Rabinovich A, Kun E. Inhibition of poly (ADP -

ribose)synthetase by gene disruption or inhibition with 5-

iodo-6-amino-1,2 benzopyrone protects mice from multiple-

low-dose- streptozotocin induced diabetes. Br J Pharmacol

2001; 133  : 909-19.

45. Baeuerle P, Baltimore D. NF-kB: ten years after. Cell 1996;

87  : 13-20.

46. Yamamoto Y, Gaynor R. IêK Kinases: key regulators of the

NFkB pathway. Trends Biochem Sci 2003; 29  : 72-9.

47. Lgssiar A, Hassan M, Schott-Ohly P, Freisen N, Nicoleti F,

Trepicchi W, et al. Interleukin 11 inhibits NFkB and AP-1

activation in islets and prevents diabetes induced with

streptozotocin in mice.  Exp Biol Med (Maywood) 2004;

229  : 425-36.

48. Rehman K, Bertera S, Bottino R, Balamurugan A, Mai J,

Mi Z, et al. Protection of islets by in situ peptide mediated

transduction of the IkB kinase inhibitor Nemo binding

domain peptide.  J Biol Chem  2003; 278   : 9862-8.

49. Wu LP, Chen LH, Zhang JS, Sun L, Zhang YQ. Protective

effect of rhIL-1beta on pancreatic islets of alloxan-induced

diabetic rats. World J Gastroenterol 2004; 10  : 3353-5.

50. Federici M, Hribal M, Perego L, Ranalli M, Caradonna Z,

Perego C, et al. High glucose apoptosis in cultured human

pancreatic islets of Langerhans: A potential role for

regulation of specific Bcl family genes toward an apoptotic

cell death program.  Diabetes  2001; 50  : 1290-301.

51. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka

H, Spinas G, et al. Glucose induced  cell production of IL-1  contributes to glucotoxicity in human pancreatic islets.

 J Clin Invest  2002; 110  : 851-60.

52. El-Assad W, Buteau J, Peyot M, Nolan C, Roduit R, Hardy

S, et al. Saturated fatty acids synergize with elevated glucose

to cause pancreatic beta cell death.  Endocrinology  2003;

144  : 4154-63.

53. Piro S, Anello M, Di Pietro C, Lizzio N, Patane G,

Rabauazzo A, et al. Chronic exposure to free fatty acids or

high glucose induces apoptosis in rat pancreatic islets:

possible role of oxidative stress.  Metabolism  2002; 51  :

1340-7.

54. Kawasaki E, Abiru N, Eguchi K. Prevention of Type 1

diabetes: from the viewpoint of beta cell damage.  Diabetes

 Res Clin Pract  2004; 66  (Suppl 1): 27-32.

55. Weir GC, Bonner-Weir S. Scientific and political

impediments to successful islet transplantation.  Diabetes

1997; 46   : 1247-56.

56. Sutherland DE, Goetz FC, Chinn PL, Elick BA, Najarian

JS. Pancreas transplantation. Pediatr Clin North Am  1984;

31  : 735-50.

57. Ricordi C. Islet transplantation: a brave new world. Diabetes

2003; 52  : 1595-603.

58. Robertson RP. Islet transplantation as a treatment for

diabetes- A work in progress.  N Engl J Med   2004; 350  :

694-705.

59. Hayashi H, Inoue K, Aung T, Tun T, Yuanjun G, Wenjing

W, et al. Application of a novel b cell line MIN6 to a mesh-

reinforced polyvinyl alcohol hydrogel tube and three-layer

436 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 13/16

agarose microcapsules: an in vitro  study. Cell Transplant 

1996; 5 (5 Suppl 1): S65-9.

60. Ohgawara H, Miyazaki J, Nakagawa Y, Sato S, Karibe S,

Akaike T. Xenoimplantation using a diffusion chamber with

a -cell line (MIN6) as a bioartificial endocrine pancreas

(Bio-AEP). Cell Transplant   1996; 5 (5 Suppl 1): S71-3.

61. Spinas GA, Mandrup-Poulsen T, Molvig J, Baek L, Bendtzen

K, Dinarello CA, et al. Low concentrations of interleukin-1

stimulate and high concentrations inhibit insulin release from

isolated rat islets of Langerhans.  Acta Endocrinol (Copenh)

1986; 113  : 551-8.

62. Lee I, Wang L, Wells AD, Ye Q, Han R, Dorf ME, et al.

Blocking the monocyte chemoattractant protein-1/CCR2

chemokine pathway induces permanent survival of isletallografts through a programmed death-1 ligand-1-dependent

mechanism.  J Immunol 2003; 171  : 6929-35.

63. Fenjves ES, Ochoa MS, Gay-Rabinstein C, Molano RD,

Pileggi A, Mendez AJ. Adenoviral gene transfer of 

erythropoietin confers cytoprotection to isolated pancreatic

islets. Transplantation  2004; 77   : 13-8.

64. Riachy R, Vandewalle B, Kerr Conte J, Moerman E,

Sacchetti P, Lukowiak B, et al. 1,25-dihydroxyvitamin D3

protects RINm5F and human islet cells against cytokine-

induced apoptosis: implication of the antiapoptotic protein

A20.  Endocrinology  2002; 143  : 4809-19.

65. Contreras JL, Bilbao G, Smyth CA, Jiang XL, Eckhoff DE,

Jenkins SM, et al. Cytoprotection of pancreatic islets before

and soon after transplantation by gene transfer of the anti-

apoptotic Bcl-2 gene. Transplantation  2001; 71  : 1015-23.

66. Ferber S, Heimberg H, Brownlee M, Colton C. Surrogate

beta cells.  Diabetologia 1997; 40 (Suppl 3) : B39-43.

67. Ranuncoli A, Cautero N, Ricordi C, Masetti M, Molano RD,

Inverardi L. Islet cell transplantation: in vivo  and in vitro

functional assessment of nonhuman primate pancreatic islets.

Cell Transplant  2000; 9  : 409-14.

68. Cotterell AA, Kenyon NS. Alternatives to

immunosuppressive drugs in human islet transplantation.

Curr Diab Rep 2002; 2 : 377-82.

69. Oluwole OO, Depaz HA, Gopinathan R, Ali A, Garrovillo

M, Jin MX. Indirect allorecognition in acquired thymic

tolerance: induction of donor-specific permanent acceptance

of rat islets by adoptive transfer of allopeptide-pulsed host

myeloid and thymic dendritic cells.  Diabetes  2001; 50  :

1546-52.

70. Nikolic B, Takeuchi Y, Leykin I, Fudaba Y, Smith RN,

Sykes M. Mixed hematopoietic chimerism allows cure of 

autoimmune diabetes through allogeneic tolerance and

reversal of autoimmunity.  Diabetes  2004; 53  : 376-83.

71. Beilhack GF, Scheffold YC, Weissman IL, Taylor C,

Jerabeck L, Burge M, et al . Purified allogeneic

hematopoietic stem cell transplantation blocks diabetes

pathogenesis in NOD mice.  Diabetes 2003; 52  : 59-68.

72. Seung E, Iwakoshi N, Woda BA, Markees TG, Mordes JP,

Rossinni AA, et al. Allogeneic hematopoietic chimerism in

mice treated with sublethal myeloablation and anti-CD154

antibody: absence of graft-versus-host disease, induction of 

skin allograft tolerance, and prevention of recurrent

autoimmunity in islet-allografted NOD/Lt mice. Blood  2000;

95  : 2175-82.

73. Li H, Kaufman CL, Boggs SS, Johnson PC, Patrene KD,

Ildstad ST. Mixed allogeneic chimerism induced by a

sublethal approach prevents autoimmune diabetes and

reverses insulitis in nonobese diabetic (NOD) mice.

 J Immunol 1996; 156   : 380-8.

74. Kaufman CL, Li H, Ildstad ST. Patterns of hemopoietic

reconstitution in nonobese diabetic mice: dichotomy of 

allogeneic resistance versus competitive advantage of 

disease-resistant marrow.  J Immunol  1997; 158   : 2435-42.

75. Liang Y, Huang T, Zhang C, Todorov I, Atkinson M,Kandeel F. Donor CD8+ T cells facilitate induction of 

chimerism and tolerance without GVHD in autoimmune

NOD mice conditioned with anti-CD3 mAb.  Blood   2005;

105  : 2180-8.

76. Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system

of T cell costimulation. Annu Rev Immunol 1996; 14 : 233-

58.

77. Adams AB, Shirasugi N, Durham MM, Strobert E, Anderson

D, Rees P, et al. Calcineurin inhibitor-free CD28 blockade-

based protocol protects allogenic islets in non-human

primates.  Diabetes  2002; 51  : 265-70.

78. Wekerle T, Kurtz J, Ito H, Ronquillo JV, Dong V, Zhao G,

et al. Allogeneic bone marrow transplantation with co-

stimulatory blockade induces macrochimerism and tolerance

without cytoreductive host treatment.  Nat Med 2000; 6   :

464-9.

79. Seung E, Mordes JP, Rossini AA, Greiner DL.

Hematopoietic chimerism and central tolerance created by

peripheral-tolerance induction without myeloablative

conditioning.  J Clin Invest  2003; 112  : 795-808.

BHONDE et al: ISLETS IN DIABETES RESEARCH 437

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 14/16

80. Pearson T, Weiser P, Markees TG, Serreze DV, Wicker LS,

Peterson LB. Islet allograft survival induced by co-

stimulation blockade in NOD mice is controlled by allelic

variants of Idd3.  Diabetes 2004; 53  : 1972-8.

81. Gotoh M, Sato Y, Abe T, Kanazawa Y. New approaches for

successful islet transplantation.  J Hepatobiliary Pancreat 

Surg 2000; 7   : 358-63.

82. Ricordi C, Scharp DW, Lacy PE. Reversal of diabetes in

nude mice after transplantation offresh and 7-day-cultured

(240C) human pancreatic islets. Transplantation 1988; 45 :

994-6.

83. Stein E, Mullen Y, Benhamou PY, Watt PC, Hober C,

Watanabe Y. Reduction in immunogenicity of human islets

by 24 degrees C culture. Transplant Proc 1994; 26  : 755.

84. Benhamou PY, Stein E, Hober C, Miyamoto M, Watanabe

Y, Nomura Y. Ultraviolet light irradiation reduces human

islet immunogenicity without altering islet function.  Horm

 Metab Res 1995; 27 : 113-20.

85. Miyamoto M, Kenmochi T, Nakagawa Y, Une S, Moldovan

S, Atiya A. Immunogenicity of cryopreserved human islets.

Transplant Proc 1995; 27   : 3406-8.

86. Hardikar AA, Risbud MV, Remacle C, Reusens B, Hoet JJ,

Bhonde RR. Islet cryopreservation: improved recovery

following taurine pretreatment. Cell Transplant   2001; 10  :

247-53.

87. Grtochowiecki T, Gotoh M, Dono K, Takeda Y, Nishihara

M, Ohta Y. Pretreatment of crude pancreatic islets with

mitomycin C prolongs graft survival time in xenogeneic rat-

to-mouse model. Transplantation  1999; 67   : 1474-7.

88. Zeng Y, Gage A, Montag A, Rothlein R, Thistlethwaite JR,

Bluestone JA. Inhibition of transplant rejection by

pretreatment of xenogeneic pancreatic islet cells with anti-

ICAM-1 antibodies. Transplantation  1994; 58   : 681-9.

89. Takeda Y, Gotoh M, Dono K, Nishihara M, Grochowiecki

T, Kimura F. Protection of islet allografts transplanted

together with Fas ligand expressing testicular allografts.

 Diabetologia  1998; 41  : 315-21.

90. Weir GC, Quickel RR, Yoon KH, Tatarkiewicz K, Ulrich

TR, Hollister-Lock J., et al. Porcine neonatal pancreatic cell

clusters (NPCCs): a potential source of tissue for islet

transplantation.  Ann Transplant   1997; 2  : 63-8.

91. Ricordi C, Socci C, Davalli AM, Staudacher C, Vertova A,

Baro P., et al. Swine islet isolation and transplantation.

 Horm Metab Res Suppl. 1990; 25 : 26-30.

92. Garkavenko O, Croxson MC, Irgang M, Karlas A, Denner

J, Elliott RB. Monitoring for presence of potentially xenotic

viruses in recipients of pig islet xenotransplantation.  J Cl in

 Microbiol  2004; 42  : 5353-6.

93. Noel J, Rabinovitch A, Olson L, Kyriakides G, Miller J,

Mintz DH. A method for large-scale, high-yield isolation

of canine pancreatic islets of Langerhans.  Metabolism

1982; 31  : 184-7.

94. Lanza RP, Butler DH, Borland KM, Harvey JM, Faustman

DL, Solomon BA, et al. Successful xenotransplantation of 

a diffusion-based biohybrid artificial pancreas: a study using

canine, bovine, and porcine islets. Transplant Proc  1992;

24  : 669-71.

95. Eloy R, Haffen K, Kedinger M, Grenier JF. Chick embryopancreatic transplants reverse experimental diabetes of rats.

 J Clin Invest  1979; 64 : 361-73.

96. Giannarelli R, Marchetti P, Bianchi F, Villani G, Cruschelli

L, Bernardini N, et al. Isolation of islets of Langerhans from

the chicken pancreas. Transplant Proc 1994; 26   : 1127-9.

97. Coddington DA, Lawen JG, Yang H, O’Hali W, Wright JR.

Xenotransplantation of fish islets into the non-cryptorchid

testis. Transplant Proc  1997; 29  : 2083-5.

98. O’Hali W, Yang H, Pohajdak B, La Prairie A, Gross M,

Wright JR. Cryopreservation of principal islets of teleost

fish: the effect on function and islet xenograft survival.

Transplant Proc 1997; 29 : 1990-1.

99. Sun YL, Ma X, Zhou D, Vacek I, Sun AM. Porcine

pancreatic islets: isolation, microencapsulation, and

xenotransplantation.  Artif Organs  1993; 17   : 727-33.

100. O’Shea GM, Goosen MF , Sun AM. Prolonged survival of 

transplanted islets of Langerhans encapsulated in a

biocompatible membrane. Biochim Biophys Acta 1984; 804:

133-6.

101. Tatarkiewicz K, Sitarek E, Sabat M, Orlowski T. Reversal

of hyperglycemia in streptozotocin diabetic mice by

xenotransplantation of microencapsulated rat islets.

 Ann Transplant  1997; 2 : 20-3.

102. Lim F, Sun AM. Microencapsulated islets as bioartificial

pancreas. Science 1980; 210: 908-10.

103. Krestow M, Lum ZP, Tai IT, Sun A. Xenotransplantation

of microencapsulated fetal rat islets. Transplantation 1991;

51  : 651-5.

438 INDIAN J MED RES, MARCH 2007

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 15/16

104. Cochrum K, Jemtrud S, Dorian R. Successful xenografts

in mice with microencapsulated rat and dog islets.

Transplant Proc  1995; 27   : 3297-4201.

105.Balamurugan AN, Remington EJ, Ramakrishna B,

Gunasekaran S. Xenographic survival and function of 

transplanted monkey pancreatic islets in rats. Transplant 

Proc 2000; 32  : 1068-70.

106.Hamaguchi K, Tatsumoto N, Fujii S, Okeda T, Nakamura

M, Yamaguchi K, Microencapsulation of pancreatic islets.

A technique and its application to culture and

transplantation.  Diabetes Res Clin Pract  1986; 2 : 337-45.

107.Hamelmann W, Ozasa T, Cairns T, Welsh K, Gray DW.

Xenotransplantation of islets across a strong species barrierrabbit to cynomolgus monkey. Transplant Proc 1994; 26  :

1097-9.

108. Risbud MV, Bhonde RR. Islet immunoisolation: experience

with biopolymers.  J Biomater Sci Polym Ed   2001; 12  :

1243-52.

109.Risbud MV, Bhargava S, Bhonde RR.  In viv o

biocompatibility evaluation of cellulose macrocapsules for

islet immunoisolation: Implications of low molecular

weight cut-off. J Biomed Mater Res 2003; 66   : 86-92.

110.Risbud MV, Bhonde RR. Suitability of cellulose molecular

dialysis membrane for bioartificial pancreas: in vitro

biocompatibility studies.  J Biomed Mater Res  2001; 54  :

436-44.

111.George S, Nair PD, Risbud MV, Bhonde RR. Nonporous

polyurethane membranes as islet immunoisolation matrices-

biocompatibility studies.  J Biomater Appl   2002; 16   :

327-40.

112.Risbud MV, Bhonde MR, Bhonde RR. Effect of chitosan-

polyvinyl pyrrolidone hydrogel on proliferation andcytokine expression of endothelial cells: implications in

islet immunoisolation.  J Biomed Mater Res   2001; 57   :

300-5.

113.Hardikar AA, Risbud MV, Bhonde RR. A simple

microcapsule generator for islet encapsulation.

 J Biosci 1999; 24 : 371-6.

114.Charles K, Harland RC, Ching D, Opara EC. Storage and

microencapsulation of islets for transplantation. Cell

Transplant   2000; 9  : 33-8.

115.Yang H, O’Hali W, Kearns H, Wright JR. Long-term

function of fish islet xenografts in mice by alginate

encapsulation. Transplantation  1997; 64  : 28-32.

116. Iwata H, Kobayashi K, Takagi T, Oka T, Yang H, Amemiya

H, et al. Feasibility of agarose microbeads with xenogeneic

islets as a bioartificial pancreas.  J Biomed Mater Res

1994; 28 : 1003-11.

117. Lhommeau C, Toillon S, Pith T, Kessler L, Jesser C, Pinget

M. Polyamide 4, 6 membranes for the encapsulation of 

Langerhans islets: preparation, physico-chemical properties

and biocompatibility studies.  J Mater Sci Mater Med 

1997; 8 : 163-74.

118. Cruise GM, Hegre OD, Lamberti FV, Hager SR, Hill R,

Scharp DS, et al. In vitro and in vivo performance of porcineislets encapsulated in interfacially photopolymerized poly

(ethylene glycol) diacrylate membranes. Cell Transplant 

1999; 8   : 293-306.

119. Zekorn T, Siebers U, Filip L, Mauer K, Schmitt U, Bretzel

RG, et al. Bioartificial pancreas: the use of different hollow

fibers as a diffusion chamber. Transplant Proc 1989; 21 :

2748-50.

120.Prevost P, Flori S, Collier C, Muscat E, Rolland E.

Application of AN69 hydrogel to islet encapsulation.

Evaluation in streptozotocin-induced diabetic rat model.

 Ann NY Acad Sci 1997; 831 : 344-9.

121.Soldani G, Giusti P, Marchetti P. Polyurethrane-

Polydimethylsiloxane (PU-PDMS) tubular membranes for

pancreatic islet transplantation: perm selectivity and

diffusion studies. J Mater Sci Mater Med  1992; 3 : 371-6.

122.Zondervan GJ, Hoppen HJ, Pennings AJ, Fritschy W,

Wolters G, Van Schilfgaarde R. Design of a polyurethane

membrane for the encapsulation of islets of Langerhans.

 Biomaterials  1992; 13  : 136-44.

123.Bonner-Weir S. Two pathways of beta-cell growth in the

regenerating rat pancreas: Implications for islet

transplantation.  Diabetes Nutr Metab   1992; 5  : 1-3.

124.Bonner-Weir S. Perspective: postnatal pancreatic   cell

growth.  Endocrinology  2000; 141  : 1926-9.

125. Rosenberg L. Induction of islet cell neogenesis in the adult

pancreas: the partial duct obstruction model.  Microsc Res

Tech 1998; 43  : 337-46.

126. Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck AB,

Cornelius JG. Reversal of insulin-dependent diabetes using

BHONDE et al: ISLETS IN DIABETES RESEARCH 439

8/13/2019 Isolated Islets in Diabetes Research

http://slidepdf.com/reader/full/isolated-islets-in-diabetes-research 16/16

islets generated in vitro from pancreatic stem cells. Nature

 Med  2000; 6   : 278-82.

127.Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song

KH, Sharma A, et al. In vitro  cultivation of human islets

from expanded ductal tissue. Proc Natl Acad Sci USA 2000;

97   : 7999-8004.

128.Katdare MR, Bhonde RR, Parab PB. Analysis of 

morphological and functional maturation of neoislets

generated in vitro  from pancreatic ductal cells and their

suitability for islet banking and transplantation.

 J Endocrinol  2004; 182  : 105-12.

129. Banerjee M, Bhonde R. Islet generation from intra islet

precursor cells of diabetic pancreas: in vitro  studies

depicting in vivo  differentiation.  JOP 2003; 4  : 137-45.

130.Kanitkar M, Bhonde R. Existence of islet regenerating

factors within the pancreas.  Rev Diab Stud  2004; 1 : 185-

92.

131.Dor Y, Brown J, Martinez OI, Melton DA. Adult pancreatic

beta-cells are formed by self-duplication rather than stem-

cell differentiation.  Nature 2004; 429  : 41-6.

132. Swenne I. Pancreatic beta-cell growth and diabetes mellitus.

 Diabetologia  1992; 35  : 193-201.

133.Smith FS, Rosen KM, Villa-Komaro L., Weir GC, Bonner-

Weir S. Enhanced insulin-like growth factor I gene

expression in the regenerating rat pancreas. Proc Natl Acad 

Sci USA 1991; 88  : 6152-6.

134.Hardikar AA, Bhonde RR. Modulating experimental

diabetes by treatment with cytosolic extract from the

regenerating pancreas. Diabetes Res Clin Pract  1999; 46  :

203-11.

135. Swenne L, Andersson A. Effect of genetic background on

the capacity for islet cell replication in mice.  Diabetologia

1984; 27   : 464-7.

136.Sjoholm A. Glucose stimulates islet beta-cell mitogenesis

through GTP-binding proteins and by protein kinase C-

dependent mechanisms.  Diabetes   1997; 46   : 1141-7.

 Reprint requests: Dr Ramesh Ramchandra Bhonde, National Centre for Cell Science NCCS Complex

Pune University Campus, Ganeshkhind, Pune 411007, India

e-mail: [email protected]

440 INDIAN J MED RES, MARCH 2007

137.Movassat J, Beattie GM, Lopez AD, Portha B, Hayek A.

Keratinocyte growth factor and beta-cell differentiation in

human fetal pancreatic endocrine precursor cells.

 Diabetologia  2003; 46   : 822-9.

138. Oberg-Welsh C, Sandler S, Andersson A, Welsh M. Effects

of vascular endothelial growth factor on pancreatic duct

cell replication and the insulin production of fetal islet-

like cell clusters in vitro. Mol Cell Endocrinol 1997; 126  :

125-32.

139.Meneur C, Elghazi L, Czernichow P, Scharfmann R.

Epidermal growth factor increases undifferentiated

pancreatic embryonic cells in vitro: a balance between

proliferation and differentiation.  Diabetes   2001; 50   :

1571-9.

140.Sjoholm A, Welsh N, Hellerstrom C. Lithium increases

DNA replication, polyamine content, and insulin secretion

by rat pancreatic beta-cells.  Am J Physiol  1992; 262  : C

391-5.

141.King DL, Kitchen KC, Chick WL. Pancreatic beta-cell

replication: relation to insulin secretion.  Endocrinology

1978; 103  : 1321-7.

142.Movassat J, Portha B. Beta-cell growth in the neonatal

Goto-kakisaki rat and regeneration after treatment withstreptozotocin at birth.  Diabetologia   1999; 42  : 1098-

106.

143.Binette TM, Dufour JM, Korbutt GS.  In vitro   maturation

of neonatal porcine islets: a novel model for the study of 

islet development and xenotransplantation.  Ann NY Acad 

Sci 2001; 944 : 47-61.

144.Welsh M, Anneren C, Lindholm C, Kriz V, Oberg-Welsh

C. Role of tyrosine kinase signaling for beta-cell replication

and survival. Ups J Med Sci 2000; 105 : 7-15.

145.Kobinger GP, Deng S, Louboutin JP, Vatamaniuk M,

Rivera VM, Lian MM, et al. Pharmacologically regulated

regeneration of functional human pancreatic islets.  Mol

Ther  2005; 11  : 105-11.


Recommended