ArticlePDF Available

Pancreatic islet cell transplantation

Authors:
  • Institute of Experimental Medicine

Abstract

Type 1 Diabetes mellitus is an autoimmune disease characterized by the destruction of insulin-producing beta cells in the pancreas. These patients become dependent on external insulin because they cannot produce insulin. However, in the long-term, exogenous insulin treatment does not prevent diabetic complications. In order to find an alternative diabetic treatment, clinical studies have demonstrated that pancreas transplantation or purified pancreatic islet transplantation could maintain control over blood sugar levels in Type 1 diabetes patients and were more effective treatment methods for these patients. Pancreatic islet transplantation is carried out with a smaller transplantation mass compared to complete pancreas transplantation. This also allows a more minor operation and transplantation of less immunologic tissue. Therefore, new methods aim to improve pancreatic islet transplantation.
doi: 10.5606/dsufnjt.2019.003
Demiroglu Science University Florence Nightingale Journal of Transplantation 2019;4(1-2):16-22
Pancreatic islet cell transplantation
Mine Ün1, Oytun Erb2
İletişim adresi: Mine Ün. İstanbul Aydın Üniversitesi Tıp Fakültesi, 34295 Sefaköy, Küçükçek mece, İstanbul, Türkiye.
e-posta: unmine13@gmail.com
ABSTRACT
Type 1 Diabetes mellitus is an autoimmune disease characterized by the destruction of insulin-producing beta cells in the pancreas. These patients
become dep endent on external in sulin because they cannot p roduce insulin. However, in the long -term, exogenous insu lin treatment does not preven t
diabetic complications. In order to find an alternative diabetic treatment, clinical studies have demonstrated that pancreas transplantation or purified
pancreatic isl et transplantation could maintain control over blood sugar levels in Type 1 diabetes patients and were more ef fective treatment methods
for these patients. Pancreatic islet transplantation is carried out with a smaller transplantation mass compared to complete pancreas transplantation.
This also allows a more minor operation and transplantation of less immunologic tissue. Therefore, new methods aim to improve pancreatic islet
transplantation.
Keywords: Beta cells, pancreatic islet transplantation, type 1 diabetes mellitus.
INSULIN HORMONE
Energy is the body’s most basic necessity in
order to maintain life. Through a hormone called
insulin produced by beta cells in the pancreas,
sugar (glucose) acquired from ingested food or
excreted to the blood from the stores in the liver
enters cells and is converted into the energy
required for the body.[1-3]
Blood sugar decreases when it enters cells.
That is, insulin is the main hormone to decrease
blood sugar. The beta cells of the pancreas
continually control blood sugar and release insulin
when the body needs it.[1- 3]
TYPE 1 DIABETES MELLITUS
The autoimmune disease in which the
pancreatic Langerhans islet beta cells are deemed
foreign and eliminated by the body’s defense
cells is called type 1 diabetes mellitus. In type 1
diabetes, destruction of beta cells renders the body
unable to produce insulin and becomes dependent
on external insulin intake.[4,5]
Type 1 diabetes is a disease of increasing
prevalence which reduces quality of life and life
expectancy and may threaten vital organs with
complications such as chronic hyperglycemia,
nephropathy, retinopathy, and neuropathy.[4-8]
In order to maintain blood sugar levels, patients
with type 1 diabetes require external insulin
intake. Banting and Best’s discovery of insulin
in the early 1920s was revolutionary. Diabetes,
which previously caused rapid mortality, became
preventable with this discovery.[7-10 ]
Insulin injection is used to delay or even
prevent the occurrence of complications such
as nephropathy, neuropathy, and retinopathy
in the short-term, however, in the long-term,
these patients may develop chronic diabetic
complications (nonhealing wounds, renal failure,
blindness, cardiovascular diseases, etc.) which
worsen over time.[4 ,5 ,7, 11]
Cite this article as:
Ün M, Erbaş O. Pancreatic islet cell transplantation. D J Tx Sci 2019;4(1-2):16-22.
1Istanbul Aydın University Medical Faculty, Istanbul, Turkey
2Department of Physiology, Demiroğlu Bilim University Faculty of Medicine, Istanbul, Turkey
17
Pancreatic islet cell transplantation
In other words, external insulin intake is
inadequate. Clinical studies have shown that
pancreas or purified pancreas islet transplantation
may ensure blood sugar control in type 1 diabetes
patients and are more effective treatment
methods.[12-14]
PANCREAS TRANSPLANT
Pancreas transplantation is the most effective
form of treatment in type 1 diabetes. However,
pancreas transplantation is a major surgical
procedure. It is also difficult to find a compatible
pancreas, and in order to prevent organ rejection,
recipients must use immunosuppressant’s for the
continuation of their lives, which has relatively
harmful effects. Therefore, pancreas transplantation
is only performed on end stage diabetic patients
who have developed type 1 diabetes-related renal
failure or other serious complications.[12, 14]
PANCREAS ISLET (BETA CELL)
TRANSPLANTATION
The best alternative to pancreas organ
transplantation in type 1 diabetes is pancreas islet
transplantation.[7,13,15]
Since Langerhans islets in the pancreas contain
insulin-producing beta cells, transplantation
of these islets alone is a relatively interesting
approach to type 1 diabetes. Islets account for
only 1-2% of the pancreas, and is therefore a
much smaller transplantation mass compared to
complete pancreas transplant. This allows for a
more minor surgical operation and transplantation
of less immunological tissue.[7,13,15 ]
Another advantage of islet transplant is that
healthy islets can be isolated from donor pancreas
even when the entire pancreas is unsuitable for
transplantation.[12,15 ]
ISLET TRANSPLANT STUDIES
For the first time in 1967, Paul Lacy developed
a new collagenase-based method for pancreas
islet isolation in rats. The method was further
modified by Dr. Camillo Ricordi, who worked
together with Lacy to successfully isolate islets in
in vitro and in vivo experiments.[16-29]
The first successful animal models for
successful islet transplantation was published in
the 1960s-1970s. Studies demonstrated how the
transplanted islets prevented diabetes in both
rodents and nonhuman primates.[19,20,28,29]
In the 1970s and 1980s, successful animal
models were attempted in humans. However,
since pancreas islets are structurally different in
humans compared to small animals (rats, guinea
pigs, etc.), methods used to isolate islet cells
in small cells were ineffective when applied in
humans. Therefore, these methods were aimed to
be adapted for humans.[21,28,29]
In the mid-1980s, the first human islet
transplantation was performed. Initial attempts
at islet transplantation had relatively low
success rates. According to studies, transplanted
islets were able to prevent diabetes after one
year of insulin synthesis in only one out of
10 patients.[7,21,22,28,29]
In the late 1990s, despite developments
with continuing transplants, only about 10% of
recipients were able to achieve normal blood
sugar (euglycemia).[7,22,28,29]
In later years, yearly islet transplantation
success rates leaped from 10 to 50% with
advancements in islet isolation and purification
techniques. In other words, at least half of patients
who underwent islet transplantation achieved
insulin independence in over a year.[7,21,22,28,29]
In following years, more successful islet
transplantations were performed. The most
important of these transplantations were
performed in 2000 by Dr. James Shapiro et al.[22]
at the Alberta University Diabetes Institute in
Edmonton, Canada. By using islets from multiple
donors, transplantation was conducted in seven
type 1 diabetes patients and all seven patients
achieved normal blood sugar levels without
exogenous insulin use over the period of one year.
In this manner, annual insulin independence rate
attained 100% with the Edmonton method.[22,28]
By introducing the world to the clinical
method to be known as the Edmonton protocol,
James Shapiro et al.[22] achieved a revolutionary
achievement in islet transplantation studies.[28]
This protocol was adapted by islet
transplantation centers worldwide and
greatly increased the success rates of islet
transplantation.[7,28]
D J Tx Sci18
HOW ARE ISLETS TRANSPLANTED?
The pancreas required for pancreas islet
transplant is obtained from persons who have
undergone brain death and islet cells are obtained
during a 6 to 8-hour long process of enzymatic
degradation of the pancreas and extraction of
beta cells. Currently, relatively pure islet cell
preparations can be obtained with collagenase
digestion of the pancreas, followed by the use
of Ficoll-Conray discontinuous gradient for the
separation of isolated islets.[13,28,29]
With many special drug combinations that
prevent organ rejection (sirolimus, tacrolimus,
daclizumab), isolated beta cells are transferred to
the liver through a catheter attached to the portal
vein (the main vessel that carries blood from the
intestines to the liver) under local anesthesia. The
islets of insulin that are transported to the liver
begin to secrete insulin to regulate blood sugar
(Figure 1).[7,28]
Although islet transplantation to the liver
(Edmonton method) may seem like it would have
long-term effect, studies by the same authors
showed that islets transplanted to the liver lost
function within the first few years, and most
patients require repeated injections after five
years.[ 7,2 3 ]
Studies have leaned towards comprehending
why islets lose function in the liver. After
administration to the portal vein, the islets reach
the liver and must adapt to it, however, they are
faced with negative conditions. Immediately after
administration to the vein, islets are exposed to
higher levels of drugs and nutrients such as glucose
in the portal system compared to the peripheral
circulation, causing loss of islet function.[24]
It should also be known that
immunosuppressant drugs used by the recipients
are another one of the main causes of function
loss in transplanted islets.[7,23,24]
DISADVANTAGES OF ISLET
TRANSPLANTATION
Although islet transplantation seems easy,
there are serious difficulties in isolating beta cells
from the pancreas without harming islets. In
addition, a considerable number of islets must be
transplanted in order to maintain blood glucose
levels for a long period of time. Since islets cannot
be extracted without completely harming all of
them, a single transplantation requires at least
two human donor pancreases. Furthermore, islet
transplantations must be repeated more than
once.[7,25,26]
Even if a large number of islets are successfully
transplanted, the recipient’s immune system
attacks the transplanted beta cells. Therefore,
the patient’s immune system must be suppressed.
Figure 1. Edmonton method. Beta cells isolated from the donor’s pancreas are injected to the recipient’s
portal vein. These cells settle in the liver and begin to produce insulin.[7]
19
Pancreatic islet cell transplantation
As in all transplantation procedures, recipients
are required to use immunosuppressants for
the rest of their life after islet transplantation.
High doses of immunosuppressive drugs are also
harmful (Figure 2).[7,27-29]
Therefore, it is necessary to develop new
methods to protect the transplanted islets from
prolonged destruction in the body and keep them
alive longer without the use of immunosuppressive
drugs which have the potential to cause serious
side effects in patients.[30]
NEW METHODS TO INCREASE ISLET
CELL LONGETIVITY
There have been significant developments in
islet transplantation over the last 10 years.[31-33 ]
It has been understood that one of the reasons
transplanted cells undergo rapid destruction
with the Edmonton method is that they are
transplanted to the liver, which contains high
levels of toxic drugs. Therefore, researchers have
studied alternative regions to transplant the islets.
Studies investigating transplantation into regions
such as arm muscle, spleen, renal capsule, and
bone marrow have found longer prolonged vitality
of the islets in these regions.[34-36]
In ongoing studies, it is aimed to transplant
islets without immunosuppressive drugs. In one
of the first methods developed to transplant
islets without immunosuppressive drugs, islets are
first encapsulated with materials to protect them
from external factors (encapsulation) and then
transplanted to recipients (Figure 3).[28,29,37]
These studies aim to protect the islets from
the recipient’s immune system by encapsulating
them with an encasement. However, this time,
the immune system develops tissue inflammation
against the encapsulation material. These
encapsulation studies on islet transplantation have
been unsuccessful.[28.29,37]
Researchers who realized they could not
protect the islets from the immune defense system
later attempted to transplant islets into regions
with the least amount of immune response. With
this purpose in mind, islet transplantation was
attempted in regions such as the brain, thymus,
and testes, rather than the liver. However, these
transplantations were also unsuccessful because
of the toxic molecules excreted by the molecules
of the tissue cells themselves. It was later thought
that it would be more reasonable to introduce the
corresponding molecules synthesized in these
tissues into the islets prior to transplantation by
gene transfer instead of transferring the islets to
these regions.[28,29,38]
After the idea of gene transplants to the islets,
the human gene TRAIL (TNF Related Apoptosis
Figure 2. Disadvantages of islet transplantation and methods of prevention.[27]
D J Tx Sci20
Figure 3. Encapsulation. Islets are transplanted to the
recipient after being encapsulated by materials capable of
protecting them from the recipient’s immune system.[28]
Inducing Ligand) came to mind. This gene has
the ability to suppress the immune system of
the recipient. With this aim, a large number
of islets were isolated from the pancreases of
rats. The TRAIL gene was transferred to islets
with adenovirus, which is a suitable vector for
gene transfer (Adenovirus is the most commonly
used gene vector in clinical gene therapy trials
today).[39 -48]
Later, two groups of rats with Streptozotocin-
induced type 1 diabetes were surgically
transplanted with islets with and without gene
transfer under their renal capsules. Rats with islet
transplantations without gene transfer had normal
blood sugar levels for a short period of time which
later reelevated. Increased blood sugar showed that
the transplanted islets were eventually rejected.
Islet transplantation after gene transfer, however,
maintained normal blood sugar levels for a longer
period of time compared to transplantation without
gene transfer. In addition, renal sections of rats
with gene-transferred islets showed a significantly
lower rate of immune cells compared to those
without gene transfer. This study demonstrated
that transferred genes protected the islets from
the recipient’s immune system and that none of
these rats were administered immunosuppressive
drugs.[43 -48] That study prevented gene-transferred
transplanted islets from being damaged by the
recipient’s immune system. This method only
provides protection to islets in the transplantation
region with gene transfer.[43-48]
In conclusion, studies on pancreas islet
transplantation are rapidly advancing and this
suggests that significant findings on this subject
will be discovered in the future.[43 -49]
Declaration of conflicting interests
The authors declared no conflicts of interest with
respect to the authorship and/or publication of this article.
Funding
The authors received no financial support for the
research and/or authorship of this article.
REFERENCES
1. Plum L, Belgardt BF, Brüning JC. Central insulin
action in energy and glucose homeostasis. J Clin
Invest 2006;116:1761-6.
2. Matthews DR, Hosker JP, Rudenski AS, Naylor
BA, Treacher DF, Turner RC. Homeostasis model
assessment: insulin resistance and beta-cell function
from fasting plasma glucose and insulin concentrations
in man. Diabetologia 1985;28:412-9.
3. Gerozissis K. Brain insulin, energy and glucose
homeostasis; genes, environment and metabolic
pathologies. Eur J Pharmacol 2008;585:38-49.
4. Kerner W, Brückel J. Definition, klassifikation und
diagnostik des diabetes mellitus. Diabetologie und
Stoffwechsel 2010;5(S 02):S109-S12.
5. Klöppel G, Löhr M, Habich K, Oberholzer M, Heitz
PU. Islet pathology and the pathogenesis of type 1
and type 2 diabetes mellitus revisited. Surv Synth
Pathol Res 1985;4:110-25.
6. Wild S, Roglic G, Green A, Sicree R, King H.
Global prevalence of diabetes: estimates for the
year 2000 and projections for 2030. Diabetes Care
2004;27:1047-53.
7. Naftanel MA, Harlan DM. Pancreatic islet
transplantation. PLoS Med. 2004;1:e58.
8. Diabetes Control and Complications Trial Research
Group, Nathan DM, Genuth S, Lachin J, Cleary P,
Crofford O, et al. The effect of intensive treatment
of diabetes on the development and progression of
long-term complications in insulin-dependent diabetes
mellitus. N Engl J Med 1993;329:977-86.
9. Epidemiology of Diabetes Interventions and
Complications (EDIC). Design, implementation, and
preliminary results of a long-term follow-up of the
Diabetes Control and Complications Trial cohort.
Diabetes Care 1999;22:99-111.
10. Writing Team for the Diabetes Control and
Complications Trial/Epidemiology of Diabetes
Interventions and Complications Research Group.
21
Pancreatic islet cell transplantation
Sustained effect of intensive treatment of type 1
diabetes mellitus on development and progression of
diabetic nephropathy: the Epidemiology of Diabetes
Interventions and Complications (EDIC) study. JAMA
2003;290:2159-67.
11. Lim DJ, Antipenko SV, Anderson JM, Jaimes KF,
Viera L, Stephen BR, et al. Enhanced rat islet function
and survival in vitro using a biomimetic self-assembled
nanomatrix gel. Tissue Eng Part A 2011;17:399-406.
12. Poggioli R, Faradji RN, Ponte G, Betancourt A,
Messinger S, Baidal DA, et al. Quality of life after islet
transplantation. Am J Transplant 2006;6:371-8.
13. Sutherland DE, Matas AJ, Najarian JS. Pancreatic
islet cell transplantation. Surg Clin North Am
1978;58:365-82.
14. Venstrom JM, McBride MA, Rother KI, Hirshberg
B, Orchard TJ, Harlan DM. Survival after pancreas
transplantation in patients with diabetes and preserved
kidney function. JAMA 2003;290:2817-23.
15. Shapiro AM, Pokrywczynska M, Ricordi C. Clinical
pancreatic islet transplantation. Nat Rev Endocrinol
2017;13: 2 6 8 -7 7.
16. Lacy PE, Kostianovsky M. Method for the isolation
of intact islets of Langerhans from the rat pancreas.
Di abete s 1967;16:35 -9.
17. Ballinger WF, Lacy PE. Transplantation of intact
pancreatic islets in rats. Surgery 1972;72:175-86.
18. Lillehei RC, Idezuki Y, Kelly WD, Najarian JS, Merkel
FK, Goetz FC. Transplantation of the intestine and
pancreas. Transplant Proc 1969;1:230-8.
19. Kemp CB, Knight MJ, Scharp DW, Lacy PE, Ballinger
WF. Transplantation of isolated pancreatic islets into
the portal vein of diabetic rats. Nature 1973;244:447.
20. Scharp DW, Murphy JJ, Newton WT, Ballinger WF,
Lacy PE. Transplantation of islets of Langerhans in
diabetic rhesus monkeys. Surgery 1975;77:100-5.
21. Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW.
Automated method for isolation of human pancreatic
islets. Diabetes 1988;37:413-20.
22. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS,
Toth E, Warnock GL, et al. Islet transplantation in
seven patients with type 1 diabetes mellitus using a
glucocorticoid-free immunosuppressive regimen. N
Engl J Med 2000;343:230-8.
23. Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E,
Kneteman NM, et al. Five-year follow-up after clinical
islet transplantation. Diabetes 2005;54:2060-9.
24. Harlan DM, Kenyon NS, Korsgren O, Roep BO;
Immunology of Diabetes Society. Current advances
and travails in islet transplantation. Diabetes
2009;58:2175-84.
25. Shapiro AM, Ricordi C, Hering BJ, Auchincloss H,
Lindblad R, Robertson RP, et al. International trial
of the Edmonton protocol for islet transplantation. N
Engl J Med 2006;355:1318-30.
26. Langer RM. Islet transplantation: lessons learned
since the Edmonton breakthrough. Transplant Proc
2010;42:1421-4.
27. Gamble A, Pepper AR, Bruni A, Shapiro AMJ.
The journey of islet cell transplantation and future
development. Islets 2018;10:80-94.
28. Farney AC, Sutherland DE, Opara EC. Evolution of
Islet Transplantation for the Last 30 Years. Pancreas
2016;45:8-20.
29. Ramírez-Domínguez M. Historical Background
of Pancreatic Islet Isolation. Adv Exp Med Biol
2016;938:1-9.
30. Ryan EA, Lakey JR, Paty BW, Imes S, Korbutt GS,
Kneteman NM, et al. Successful islet transplantation:
continued insulin reserve provides long-term glycemic
control. Diabetes 2002;51:2148-57.
31. Linetsky E, Bottino R, Lehmann R, Alejandro
R, Inverardi L, Ricordi C. Improved human islet
isolation using a new enzyme blend, liberase. Diabetes
1997;4 6:1120 - 3.
32. Bucher P, Mathe Z, Morel P, Bosco D, Andres
A, Kurfuest M, et al. Assessment of a novel two-
component enzyme preparation for human islet
isolation and transplantation. Transplantation
2005;79: 91-7.
33. Emamaullee JA, Stanton L, Schur C, Shapiro AM.
Caspase inhibitor therapy enhances marginal mass
islet graft survival and preserves long-term function in
islet transplantation. Diabetes 2007;56:1289-98.
34. Merani S, Toso C, Emamaullee J, Shapiro AM. Optimal
implantation site for pancreatic islet transplantation.
Br J Surg 2008;95:1449-61.
35. Daly B, O’Kelly K, K lassen D. Interventiona l procedures
in whole organ and islet cell pancreas transplantation.
Semin Intervent Radiol 2004;21:335-43.
36. Campbell PM, Senior PA, Salam A, Labranche
K, Bigam DL, Kneteman NM, et al. High risk of
sensitization after failed islet transplantation. Am J
Tran s plan t 20 07;7:2311-7.
37. Johansson U, Rasmusson I, Niclou SP, Forslund N,
Gustavsson L, Nilsson B, et al. Formation of composite
endothelial cell-mesenchymal stem cell islets: a novel
approach to promote islet revascularization. Diabetes.
2008;57:2393-401.
38. Sanlioglu AD, Griffith TS, Omer A, Dirice E, Sari R,
Altunbas HA, et al. Molecular mechanisms of death
ligand-mediated immune modulation: a gene therapy
model to prolong islet survival in type 1 diabetes. J
Ce ll Bio chem 200 8;10 4:710 -20.
39. Sanlioglu AD, Dirice E, Elpek O, Korcum AF, Balci
MK, Omer A, et al. High levels of endogenous tumor
necrosis factor-related apoptosis-inducing ligand
expression correlate with increased cell death in
human pancreas. Pancreas 2008;36:385-93.
40. Sanlioglu AD, Dirice E, Elpek O, Korcum AF,
Ozdogan M, Suleymanlar I, et al. High TRAIL death
receptor 4 and decoy receptor 2 expression correlates
with significant cell death in pancreatic ductal
adenocarcinoma patients. Pancreas 2009;38:154-60.
41. Holoch PA, Griffith TS. TNF-related apoptosis-
inducing ligand (TRAIL): a new path to anti-cancer
D J Tx Sci22
therapies. Eur J Pharmacol 2009;625:63-72.
42. Kahraman S, Dirice E, Sanlioglu AD, Yoldas B, Bagci
H, Erkilic M, et al. In vivo fluorescence imaging is
well-suited for the monitoring of adenovirus directed
transgene expression in living organisms. Mol Imaging
Biol 2010;12:278-85.
43. Lopez-Talavera JC, Garcia-Ocaña A, Sipula I,
Takane KK, Cozar-Castellano I, Stewart AF.
Hepatocyte growth factor gene therapy for pancreatic
islets in diabetes: reducing the minimal islet transplant
mass required in a glucocorticoid-free rat model
of allogeneic portal vein islet transplantation.
Endocrinology 2004;145:467-74.
44. Dirice E, Sanlioglu AD, Kahraman S, Ozturk S, Balci
MK, Omer A, et al. Adenovirus-mediated TR AIL gene
(Ad5hTRAIL) delivery into pancreatic islets prolongs
normoglycemia in streptozotocin-induced diabetic
rats. Hum Gene Ther 2009;20:1177-89.
45. Ahrén B, Pacini G. Islet adaptation to insulin resistance:
mechanisms and implications for intervention.
Diabetes Obes Metab 2005;7:2-8.
46. Dirice E, Kahraman S, Elpek GO, Aydin C, Balci
MK, Omer A, et al. TRAIL and DcR1 expressions
are differentially regulated in the pancreatic islets of
STZ- versus CY-applied NOD mice. Exp Diabetes Res
2011;2011:625813.
47. Shapiro AM, Lakey JR. Future trends in islet
cell transplantation. Diabetes Technol Ther
2000;2:449-52.
48. Kahraman S, Dirice E, Hapil FZ, Ertosun MG, Ozturk
S, Griffith TS, et al. Tracing of islet graft survival by
way of in vivo fluorescence imaging. Diabetes Metab
Res Rev 2011;27:575-83.
49. Ahearn A J, Parekh JR, Posselt AM. I slet transpla ntation
for Type 1 diabetes: where are we now? Expert Rev
Clin Immunol 2015;11:59-68.
... This approach is applied in the treatment of T1D, where there is a lack of insulin due to the destruction of the islet cells. A whole pancreas can be transplanted, or the islet cells producing the insulin are implanted in the patient [40].The transplantation of the whole pancreas is faced with organ rejection. The patients are given immunosuppressive agents to minimize organ transplant. ...
... In the case of islet cell transplantation, only the cells producing insulin are implanted. This gives the opportunity of transplanting only healthy cells [40]. ...
Article
Full-text available
The rising burden of Diabetes mellitus (DM) globally and particularly in sub-Sahara Africa calls for alternative treatment solutions. This is because the currently available drugs for its management are limited due to undesirable adverse effects and high cost. Thus, this review explores diabetes and summarizes its treatment options, focusing mainly on medicinal plants therapy. Information on twenty-five selected medicinal plants from sub-Sahara Africa having hypoglycemic and anti-diabetic potentials was obtained via electronic search of major databases, such as Pubmed/Medline, Scopus, Google Scholar and web of science. Predominant bioactive compounds found in these plants include tannins, carpaine, terpenoids, hexadecenoic acid, luteolin, saponins, glycosides, rutin, quercetin, vindoline and kaempferol. Robust evidence indicates that these medicinal plants and their bioactive components exert their antidiabetic potentials via different mechanisms, including: regeneration of pancreatic β-cell and insulin secretion; inhibition of α-amylase, inhibition of intestinal glucose absorption and liver glucose production; antioxidative stress; limitation of glycogen degradation and gluconeogenesis; anti-inflammatory, immunoregulatory. DM imposes a tremendous burden in the region, and its prevalence is not abating; thus the rich flora of the region with known hypoglycemic and antidiabetic efficacy could be explored as a complementary therapy in its management.
Article
Full-text available
Intraportal islet transplantation has proven to be efficacious in preventing severe hypoglycemia and restoring insulin independence in selected patients with type 1 diabetes. Multiple islet infusions are often required to achieve and maintain insulin independence. Many challenges remain in clinical islet transplantation, including substantial islet cell loss early and late after islet infusion. Contributions to graft loss include the instant blood-mediated inflammatory reaction, potent host auto- and alloimmune responses, and beta cell toxicity from immunosuppressive agents. Protective strategies are being tested to circumvent several of these events including exploration of alternative transplantation sites, stem cell-derived insulin producing cell therapies, co-transplantation with mesenchymal stem cells or exploration of novel immune protective agents. Herein, we provide a brief introduction and history of islet cell transplantation, limitations associated with this procedure and methods to alleviate islet cell loss as a means to improve engraftment outcomes.
Article
Full-text available
In this article, we will review the changes that have occurred in islet transplantation at the birth of Pancreas 30 years ago. The first attempts at β-cell replacement in humans, pancreas and islet transplantation, were performed in the 1960s and 1970s. Although pancreas transplantation has been an accepted treatment for severe labile diabetes predating the emergence of the journal, allogeneic islet transplantation remains experimental. Current investigations within islet transplantation focus to improve islet function after transplantation. Improving islet viability during isolation, exploring ways to increase engraftment, and protection from the host immune system are some of the goals of these investigative efforts. The major barriers to clinical islet transplantation are shortage of human pancreas, the need for immunosuppression, and the inadequacy of the islet isolation process. It is generally accepted that islet encapsulation is an immunoisolation tool with good potential to address the first 2 of those barriers. We have therefore devoted a major part of this review to the critical factors needed to make it a clinical reality. With improved islet isolation techniques and determination of the best site of engraftment as well as improved encapsulation techniques, we hope that islet transplantation could someday achieve routine clinical use.
Article
Full-text available
Purpose: We tested a new light detection cooled charge-coupled device (CCD) for in vivo assessment of noninvasive, whole-body fluorescence optical imaging of adenovirus directed enhanced green fluorescent protein (AdEGFP) expression. Procedures: AdEGFP was injected i.v. into BALB/c mice via tail vein. Whole-body fluorescence optical imaging of AdEGFP expression was performed using a Kodak 2000MM Image Station before and after vector administration. Results: EGFP expression was exclusively detected around the abdominal cavity, and the fluorescent signal peaked at day 4 and then remained detectable for at least 30 days. Ex vivo fluorescence imaging confirmed that EGFP expression was restricted to the liver, and transgene expression was homogeneously diffused into all four lobes. Conclusions: These findings demonstrate that in vivo fluorescence imaging provides functional data indicating the approximate location, magnitude, and duration of AdEGFP expression.
Article
Full-text available
TNF-related apoptosis-inducing ligand (TRAIL) is an important component of the immune system. Although it is well acknowledged that it also has an important role in Type 1 Diabetes (T1D) development, this presumed role has not yet been clearly revealed. Streptozotocin (STZ) and Cyclophosphamide (CY) are frequently used agents for establishment or acceleration of T1D disease in experimental models, including the non-obese diabetic (NOD) mice. Although such disease models are very suitable for diabetes research, different expression patterns for various T1D-related molecules may be expected, depending on the action mechanism of the applied agent. We accelerated diabetes in female NOD mice using STZ or CY and analyzed the expression profiles of TRAIL ligand and receptors throughout disease development. TRAIL ligand expression followed a completely different pattern in STZ- versus CY-accelerated disease, displaying a prominent increase in the former, while appearing at reduced levels in the latter. Decoy receptor 1 (DcR1) expression also increased significantly in the pancreatic islets in STZ-induced disease. Specific increases observed in TRAIL ligand and DcR1 expressions may be part of a defensive strategy of the beta islets against the infiltrating leukocytes, while the immune-suppressive agent CY may partly hold down this defense, contributing further to diabetes development.
Article
Clinical pancreatic islet transplantation can be considered one of the safest and least invasive transplant procedures. Remarkable progress has occurred in both the technical aspects of islet cell processing and the outcomes of clinical islet transplantation. With >1,500 patients treated since 2000, this therapeutic strategy has moved from a curiosity to a realistic treatment option for selected patients with type 1 diabetes mellitus (that is, those with hypoglycaemia unawareness, severe hypoglycaemic episodes and glycaemic lability). This Review outlines the techniques required for human islet isolation, in vitro culture before the transplant and clinical islet transplantation, and discusses indications, optimization of recipient immunosuppression and management of adjunctive immunomodulatory and anti-inflammatory strategies. The potential risks, long-term outcomes and advances in treatment after the transplant are also discussed to further move this treatment towards becoming a more widely available option for patients with type 1 diabetes mellitus and eventually a potential cure.
Article
Pancreatic islet transplantation is a minimally invasive procedure that can restore normoglycemia and insulin independence in Type 1 diabetics without the surgical complications associated with vascularized pancreas transplantation. The advances made in this field over the past decade have dramatically improved patient outcomes, and the procedure is now transitioning from an experimental treatment to a clinical reality. Nonetheless, a number of important issues continue to hamper the success of islet transplantation and must be addressed before there is widespread clinical acceptance. These include the relative inefficiency of the islet isolation process, the progressive loss of islet function over time and the need for multiple donors to achieve insulin independence. Here, we discuss the current status of islet transplantation and examine its future as a treatment for Type 1 diabetes.
Article
Objectives: The importance of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL receptor expression in pancreatic carcinoma development is not known. To reveal the putative connection of TRAIL and TRAIL receptor expression profile to this process, we analyzed and compared the expression profile of TRAIL and its receptors in pancreatic tissues of both noncancer patients and patients with pancreatic ductal adenocarcinoma (PDAC). Methods: Thirty-one noncancer patients and 34 PDAC patients were included in the study. TRAIL and TRAIL receptor expression profiles were determined by immunohistochemistry. Annexin V binding revealed the apoptotic index in pancreas. Lastly, the tumor grade, tumor stage, tumor diameter, perineural invasion, and number of lymph node metastasis were used for comparison purposes. Results: TRAIL decoy receptor 2 (DcR2) and death receptor 4 expression were up-regulated in PDAC patients compared with noncancer patients, and the ductal cells of PDAC patients displayed significant levels of apoptosis. In addition, acinar cells from PDAC patients had higher DcR2 expression but lower death receptor 4 expression. Increased DcR2 expression was also observed in Langerhans islets of PDAC patients. Conclusions: Differential alteration of TRAIL and TRAIL receptor expression profiles in PDAC patients suggest that the TRAIL/TRAIL receptor system may play a pivotal role during pancreatic carcinoma development.