Minireviews Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2014; 20(37): 13512-13520
Published online Oct 7, 2014. doi: 10.3748/wjg.v20.i37.13512
Review of experimental attempts of islet allotransplantation in rodents: Parameters involved and viability of the procedure
Leandro Ryuchi Iuamoto, Alberto Meyer, Eleazar Chaib, Luiz Augusto Carneiro D’Albuquerque, Department of Gastroenterology, Liver and Pancreas Transplantation Surgery Unit, University of Sao Paulo, Sao Paulo 05403-090, Brazil
Author contributions: Iuamoto LR and Meyer A read all the articles, selected the most relevant of them and edited the manuscript; Chaib E and D’Albuquerque LAC designed the study and were also involved in editing the manuscript; all authors contributed to the manuscript.
Correspondence to: Dr. Leandro Ryuchi Iuamoto, Department of Gastroenterology, Liver and Pancreas Transplantation Surgery Unit, University of Sao Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, Sao Paulo 05403-090, Brazil. leandro.iuamoto@gmail.com
Telephone: +55-11-30618322 Fax: +55-11-30618322
Received: March 7, 2014
Revised: May 3, 2014
Accepted: June 21, 2014
Published online: October 7, 2014

Abstract

The purpose of the present study was to organize the parameters involved in experimental allotransplantation in rodents to elaborate the most suitable model to supply the scarcity of islet donors. We used the PubMed database to systematically search for published articles containing the keywords “rodent islet transplantation” to review. We included studies that involved allotransplantation experiments with rodents’ islets, and we reviewed the reference lists from the eligible publications that were retrieved. We excluded articles related to isotransplantation, autotransplantation and xenotransplantation, i.e., transplantation in other species. A total of 25 studies related to allotransplantation were selected for systematic review based on their relevance and updated data. Allotransplantation in rodents is promising and continues to develop. Survival rates of allografts have increased with the discovery of new immunosuppressive drugs and the use of different graft sites. These successes suggest that islet transplantation is a promising method to overcome the scarcity of islet donors and advance the treatment options for type 1 diabetes.

Key Words: Islet transplantation, Allograft, Immunosuppression, Type 1 diabetes, Islet grafts, Diabetes mellitus, Islet, Hyperglycemia

Core tip: This is an important systematic review for readers to analyze the different existing methodologies of islet allotransplantation. This article reviews all aspects of donors and recipients, the types and dosages of immunosuppressive therapy, graft survival time and evolution of the recipient’s blood glucose. Therefore, the present article permits reproduction and improvement of the experiments involving islet allotransplantation in rodents to develop alternative therapies for type 1 diabetes.



INTRODUCTION

It is estimated that 4% of the world population is affected by diabetes mellitus, of which 10% have type 1 diabetes[1]. Furthermore, the incidence of diabetes cases in Europe has increased, especially in children and teenagers, in whom the incidence of type 1 diabetes increased by 4% last year. One trend is the occurrence of type 1 diabetes mellitus at younger ages, between 10 and 14 years. Today, the disease is already at 0-5 years[2]. According to the IBGE-CENSUS-2010, there are currently 12054827 diabetics patients in Brazil[3]. Thus, approximately 1.2 million diabetics in Brazil may benefit from research aimed at improving treatment of type 1 diabetes.

Currently, insulin is the primary treatment method for diabetes. However, approximately 5%-10% of patients have severe and unexpected fluctuations in their blood glucose levels, resulting in multiple episodes of hypoglycemia, which has serious clinical consequences. In such cases, pancreas transplantation is an alternative treatment option that is already in clinical use. Another alternative option is islet transplantation, which is a less invasive therapeutic method but is still in development[3]. Regarding the effectiveness of treatment, some results showed 80% insulin independence within the first year in postoperative patients treated with islet transplantation[4]; however, the survival rate of islets remains low.

The scarcity of islets is a significant obstacle hindering the widespread use of islet allograft therapy. According to the Network of Organ Procurement and Transplantation, in 2011, only 1562 pancreases were recovered from 8000 donor organs available in the United States. Furthermore, many donated pancreases are not suitable for islet extraction or do not fit the selection criteria. It is also common for islets to be handled incorrectly. For these reasons, only a small number of islet transplantations can be carried out[5].

Some restrictions were found in the technical development of islet transplantation: the number of donor pancreases available for islet transplantation, below that required for healing the millions of people with type 1 diabetes; technical difficulties and the cost of islet isolation; poor durability of insulin independence; and autoimmunity and rejection after transplantation, which must still be overcome. It is therefore essential to develop an unlimited source of cells capable of secreting insulin in response to glucose and that can be transplanted with little or no need for systemic immunosuppression[6,7].

The purpose of the present study is to review experimental allotransplantation procedures that have been attempted in rodents to analyze the parameters involved and the viability of the procedure.

SEARCH PROCESS
Search process

The study was performed using the PubMed database to search for published articles containing the keywords “rodent islet transplantation”. However, to filter the results, we searched PubMed records for the period January 2000-December 2013 using the following search terms for islet allotransplantation in rodents: “{rodent islet transplantation AND [‘‘2000’’(Date-Completion): ‘‘3000’’(Date-Completion) AND (allotransplantation) NOT porcine] NOT tilapia} NOT nonhuman primate”.

This ensured that articles discussing transplantation in porcine, tilapia and nonhuman primates (more common species used for transplantation) were excluded from the review to focus on those articles related to allotransplantation in rodents. Following the PubMed search, we reviewed the references from the publications retrieved and obtained the entire text of publications that could potentially be included in the systematic review. Unpublished studies and letters were ignored. Studies that did not have a full text available in English were purchased for review.

Eligible studies were selected for analysis based on the following inclusion criteria: (1) studies must be related to allotransplantation; (2) the species studied must be rodent species; and (3) articles must be relevant and the information up to date.

The review was written in English, and the relevant information, such as donor/recipient, immunosuppression, allotransplantation site, graft survival time, glucose variations and diabetes induction method, was organized into tables.

DATA ABSTRACTION

The authors abstracted the characteristics of the study, such as the source(s) for experimental (e.g., medical records and clinical databases) and relevant data, into the tables-donor/recipient; immunosuppression; allotransplantation site; graft survival time; glucose variations; and diabetes induction method.

SEARCH RESULTS

A total of 2650 articles from 2000 to 2013 were found. Only 25 articles were related to allotransplantation. These articles were selected based on their relevance and updated information (Tables 1, 2, 3, 4, 5, 6; Figure 1A and B).

Table 1 Description of the experimental studies on allografts in rodents.
Ref.Donor/recipientImmunosuppressionAllotransplantation siteGraft survival time
Fotiadis et al[9]Lewis→ WistarMycophenolate mofetil (MMF) and Cyclosporine A (CsA)SpleenN/A
Merani et al[10]Lewis →WistarAEB-071 (Protein kinase C inhibitor) + CsA, CTLA4-Ig, MMFKidney capsule100 d
Nishimura et al[11]C57BL/6 → Balb/cTacrolimusNonmetallic dorsal skinfold chamberN/A
Makhlouf et al[12]C57BL/6/Balb/cBlockade of CD28:B7 and anti-CD40L; CTLA-4Kidney capsule1 wk
Salazar-Bañuelos et al[13]Wistar →Sprague DawleyNo immunosuppressionMedullary channel21 d
Wee et al[14]Lewis → FisherCsA + Tautomycetin (synergist)Liver (portal vein)Control group - 5.2 ± 0.5 d TMC - 5.1 ± 0.9 d TMC (0.03 mg/kg) + CsA (5 mg/kg) - > 41 d TMC (0.1 mg/kg) + CsA (5 mg/kg) - 103.8 ± 56.8 d
Plesner et al[15]Balb/c →EBANo immunosuppressionKidney capsule60 d
Watanabe et al[16]Balb/c → C57BL/6Tacrolimus and DHMEQ (NF-κB inhibitor)Kidney capsule100 d
Gysemans et al[17]Balb/c→ C57BL/6No immunosuppressionKidney capsule9.2 ± 4.9 d (Autoimmune diabetes) 15 ± 3 d (Not chemically diabetic autoimmune)
Xekouki et al[18]Wistar→LewisCsA and MMFSpleen (parenchyma)8 d (CsA) 10.92 d (MMF 1) 11 d (MMF 2)
Baker et al[19]A/J→ C57Bl/6JMonoclonal antibody antiBIP-10Kidney capsule19.7 ± 2.3 d (C57Bl/6J) 20.2 ± 2.7 d (CXCR3-/-C57Bl/6J
Li et al[20]FVB→ Balb/cNo immunosuppressionKidney capsuleN/A
Vieiro et al[21]C57Bl/6→ C3HTritiated thymidine (preoperative) and CsASubcutaneousN/A
Neuzillet et al[22]C3H → Balb/cNo immunosuppressionKidney capsule13.8-27.5 d
Melzi et al[23]C57Bl/6 → Balb/cRapamycin+ FK506+ anti-IL- 2Ra chain mAbs and rapamycin+IL-10Kidney capsule> 100 d
Fiorina et al[24]Balb/c→ C57Bl/6No immunosuppressionKidney capsule14 d
Fan et al[25]C57Bl/6 → Balb/cLTß R-Ig, CTLA4-Ig or LTR mAb anti mouseLTβ R-Ig- 27 d CTLA4-Ig- 55 d LTβ R-Ig+CTLA4-Ig - > 100 d LTR mAb anti mouse - 11 d
Jung et al[26]Balb/c → C57Bl/6CD154 mAb (MR1) anti mouse and ROS-A (Reactive Oxygen Specie - A)Kidney capsuleROS-A - 53 d MR1 - 82 d ROS-A+MR1 - > 160 d
Påhlman et al[27]Balb/c → C57Bl/6JAR-C117977 (10 or 30 mg/kg) or CsA 20 mg/kgKidney capsuleCsA - 16 d AR-C117977, 10 mg/kg - >100 d AR-C117977, 30 mg/kg -29,33 d
Wang et al[28]Balb/c → C57Bl/6B7-H4 and Ad-LacZKidney capsuleB7-H4 - approximately 60 d Ad-LacZ - approximately > 20 d
Chen et al[29]Sprague Dawley → LewisNo immunosuppressionIntra-abdominal8 wk
Giraud et al[30]C3H → Balb/cNo immunosuppressionKidney capsuleSCOT + PEG 20 kDa ³10 g/L - 20 d, CMRL-1066 + 1% BSA - 17.5 ± 1 d, Solution UW - 17.2 ± 0.4 d, SCOT without PEG - 14 ± 0.9 d Solution HBSS + 0.5% BSA - 14 ± 0.7 d
Qi et al[31]Wistar/Lewis → LewisNo immunosuppressionIntraperitoneal (Macroencapsulated) Kidney capsule (not macroencapsulated)24 wk (Macroencapsulated) 48 h (not macroencapsulated)
Potiron et al[32]Wistar → C57Bl/6CTLA4 Ig or CD40 IgKidney capsule24.3 ± 9.7 d
Jahr et al[33]Lewis → WistarAnti-rat antilymphocyte serumLiver (portal vein)
Table 2 Sites of islet infusion based on the literature from the PubMed database.
Sites of islet infusionLiterature from the PubMed database
Kidney capsule70%
Liver23%
Other sites17%
Table 3 Comparative analysis of the different types of rodents used and their basic characteristics-Age and Weight.
Ref.Donor/recipientAgeWeight
Fotiadis et al[9]Lewis→ WistarN/A220 g-300 g
Merani et al[10]Lewis→ WistarN/A200 g (male)/150 g (female)
Nishimura et al[11]C57BL/6 → Balb/c9-12 wk /8-12 wkN/A
Makhlouf et al[12]C57BL/6→ Balb/c6-8 wk (male)/ N/AN/A
Salazar-Bañuelos et al[13]Wistar→ Sprague DawleyN/A260-326 g
Wee et al[14]Lewis→ Fisher10-12 wkN/A
Plesner et al[15]Balb/c→ EBA8-10 wk /N/AN/A
Watanabe et al[16]Balb/c→ C57BL/610-14 wk /maleN/A
Gysemans et al[17]Balb/c→ C57BL/6(8-21 d) → (> 180 d)N/A
Xekouki et al[18]Wistar→LewisN/A/male220-300 g
Baker et al[19]A/J→ C57Bl/6J8-12 d/maleN/A
Li et al[20]FVB→ Balb/c8-12 wkN/A
Vieiro et al[21]C57Bl/6→ C3HN/AN/A
Neuzillet et al[22]C3H → Balb/cN/AN/A
Melzi et al[23]C57Bl/6 → Balb/c9 wk (female) → 9 wk (female)20-22 g
Fiorina et al[24]Balb/c→ C57Bl/6N/AN/A
Fan et al[25]C57Bl/6 → Balb/cN/A - adults (female)N/A
Jung et al[26]Balb/c → C57Bl/612 wk (male) → 12 wk (male)25-30 g
Påhlman et al[27]Balb/c → C57Bl/6JN/A - (female)N/A
Wang et al[28]Balb/c → C57Bl/68-10 wk (female) → 8-10 wkN/A
Chen et al[29]Sprague Dawley → LewisN/A (male)250-350 g → 196 ± 15 g
Giraud et al[30]C3H → Balb/c6 wkN/A
Qi et al[31]Wistar/Lewis → Lewis9-10 wk (male)250-300 g
Potiron et al[32]Wistar → C57Bl/6N/A (male)200-300 g
Jahr et al[33]Lewis → WistarN/A (male) → N/A (male)310-330 g→ 215-245 g
Table 4 Analysis of the immunosuppressant drugs used at international islet transplantation research centers.
ImmunosuppressantNumber of centers using theimmunosuppressant(based on data from the literature)
CsA6
MMF3
CTLA4 Ig4
CD40 Ig2
NF-kB Inhibitor (DHMEQ)1
Anti-CD154 mAb (MR1)2
Tritiated thymidine1
Tacrolimus1
Blockade of CD28:B71
Tautomycetin1
Protein Kinase C Inhibitor (AEB-071)1
Monoclonal antibody anti-BIP-101
Rapamycin+FK506+anti–IL-
2Ra chain mAbs, n31 and rapamycin+IL-10; n291
LTβ R-Ig1
LTR mAb1
ROS-A1
AR-C1179771
B7-H4 and Ad-LacZ1
Anti-rat antilymphocyte serum1
No immunosuppression9
Table 5 Quantitative analysis of immunosuppressant drugs use.
Ref.ImmunosuppressionDoseAdministration frequency
Fotiadis et al[9]MMF and CsA12 mg/kg and 23 mg/kg (MMF) 5 mg/kg (CsA)-
Merani et al[10]AEB-071 (Protein Kinase C Inhibitory) + CsA, CTLA4-Ig, MMF30 mg/kg (AEB-071) 2.5 mg/kg and 5 mg/kg (CsA) 0.25 mg (CTLA4-Ig Intraperitoneal) 10 mg/kg (MMF)2 times a day, oral (AEB-071) 2 times a day, oral (CsA) 0, 2, 4 and 6 PO, Intraperitoneal (CTLA4-Ig) Once a day, oral (MMF)
Nishimura et al[11]Tacrolimus0.5 mg/kgInfused subcutaneously - Daily - for 14 d
Makhlouf et al[12]Blockade of CD28:B7 and anti-CD40 L; CTLA-4250μgIntraperitoneal - 0, 2, 4 and 6 PO
Wee et al[14]CsA+Tautomycetin (Synergist)5 mg/g and 15 mg/kg (CsA)Once a day for 7 d
Watanabe et al[16]Tacrolimus and DHMEQ1.5 mg/kg (Tacrolimus) 20 mg/kg (DHMEQ)Once a day 0 to 3 PO and 2 times a day 0 to 14 PO (DHMEQ); 0 to 14 PO (Tacrolimus); Once a day 0 to 3 PO (DHMEQ)+0 to 14 PO (Tacrolimus)
Xekouki et al[18]CsA and MMF15 mg/kg (CsA) 12 mg/kg (MMF) 23 mg/kg (MMF)Oral - Daily - 12 consecutive days
Baker et al[19]Monoclonal antibody antiBIP-10300 μg intraperitonealDaily - 14 d1
Vieiro et al[21]Tritiated thymidine (preoperative) and CsA20 mg/kg (CsA)N/A
Melzi et al[23]Rapamycin + FK506 + anti–IL-2Ra chain mAbs and rapamycin+IL-101 mg/kg (Rapamycin) 0.05 μg/kg (IL-10) 0.3 mg/kg (FK506) 1 mg/kg (mAbs)Intraperitoneal: Once a day - 30 PO (Rapamycin) 2 times a day - 30 d (IL-10) Once a day - 30 d (FK506) 0.4 PO (mAbs)
Fan et al[25]LTβ R-Ig, CTLA4-Ig or LTR mAb anti mouse200 μgIntraperitoneal- days -1, 1, 3, 5, 7 and 9
Jung et al[26]CD154 mAb (MR1) anti mouse + ROS-A250 μg (CD154 mAb (MR1) anti mouse) 200 mg/kg of Ros AIntraperitoneal injection 0, 2, 4, 6 and 8 PO (CD154 mAb (MR1) anti mouse) 8 consecutive days (ROS-A)
Påhlman et al[27]AR-C117977 or CsA0.2 mL - 3, 10, 30, or 100 mg/kg (AR-C117977) 0.5 mL - 20 mg/kg (CsA)Subcutaneous - once a day 0 to 9 PO (AR-C117977) Once a day 0-9 PO or 0-39 PO (CsA)
Wang et al[28]B7-H45 plaque-forming units (pfu) of Ad-B7-H4 or Ad-LacZN/A
Potiron et al[32]CTLA4 Ig or CD40 Ig5 109 IP of AdCTLA4 IM and/or 5 109 IM or 2 109 IV of AdCD40Ig; IM administration: 10 μL per point (3 points) IV administration: 150 μL with 0.9% sodium chlorideIM administration - anterior tibialis muscle; IV administration - venile vein
Jahr et al[33]Anti-rat antilymphocyte serumIntraperitoneal administration 0.5 mL1 d after islet transplantation
Table 6 Analysis of induction and treatment of diabetic process with islet transplantation.
Ref.Number oftransplanted isletsDiabetes induction methodHyperglycemia induction (preoperative)Normalization of hyperglycemia (postoperative)Graft rejectionCriteria for primary graft dysfunction (PGD)
Fotiadis et al[9]1812 ± 145Streptozotocin (60 mg/kg) + PBS-Solution (Phosphate Buffer Solution) - 10 mg/mL (pH 4,5);7 d3 d12 d (MMF) 10 d (CsA)Glucose above 200 mg/dL; after 2nd PO 2 consecutive times
Merani et al[10]1500Streptozotocin (75 mg/kg) intraperitoneal5 d3 d22 dGlucose above 324 mg/dL after 2 consecutive days
Nishimura et al[11]2-10/dorsal skinfold chamber---N/A-
Makhlouf et al[12]350 (Balb/c) 700 (NOD)Streptozotocin and spontaneously (225 mg/kg in peritoneal cavity)2 wk3 d10 d (Balb/c)200 mg/dL - 2 to 3 consecutive days
5 d (NOD) and 7 d complete rejection (NOD)
Salazar-Bañuelos et al[13]840 (of Wistar)---N/A1N/A
Wee et al[14]4000Streptozotocin (35 mg/kg)N/AN/AUntreated - 5.2 d (± 0.5)200 mg/dL after 2 consecutive days
TMC - 5, 1 d (± 0, 9)
TMC (0.03 mg/kg) + CsA (5 mg/kg) - > 41 d
TMC (0.1 mg/kg) + CsA (5 mg/kg) - 103.8 ± 56.8 d
Plesner et al[15]550Streptozotocin (375 mg/dL) intraperitoneal3-5 d5 d160 d≥ 198 mg/dL after 2 consecutive days
Watanabe et al[16]600 or 300Streptozotocin (180 mg/kg) intraperitoneal5-7 dN/A69 d (Tacrolimus)> 350 mg/dL for 2 consecutive days
100 d (DHMEQ 3 d and Tacrolimus 14 d)
Gysemans et al[17]300Alloxan (90 mg/kg)24 hN/AN/A - 23%Glucose level > 200 mg/dL more than 3 consecutive days
Xekouki et al[18]2000Streptozotocin (60 mg/kg) diluted in phosphated solution 10 mg/mL1 wkN/A7 d (MMF ´1)-
Baker et al[19]300Streptozotocin (220 mg/kg)N/AN/A7 d-
Li et al[20]400 (200/Kidney capsule)Streptozotocin (220 mg/kg)N/AN/A8.36 ± 1.67 (islets of FVB)Glucose levels > 250 mg/dL - 2 consecutive measurements
16.2 ± 2.52 (islets of MT)
Vieiro et al[21]200Streptozotocin (270 mg/kg) intraperitonealN/AN/A3-7 d≥ 250 mg/dL - 3 consecutive measurements
Neuzillet et al[22]550N/AN/A4 hPEG-Solution 8 kDa 27.50 ± 3.70 d; PEG PEG-Solution 20 kDa 23.13 ± 4.39 d; PEG-Solution 35 kDa 13.80 ± 3.49 d> 199.8 mmol/L - 2 consecutive measurements
Melzi et al[23]400175 a 200 mg/kg intravenous1-2 wk5 d29 d (mouse with Glucose levels < 450 mg/dL) and 16 d (mouse with Glucose levels > 450 mg/dL)> 250 mg/dL - 2 consecutive measurements on postoperative
Fiorina et al[24] Fan et al[25]NA 500StreptozotocinN/A N/AN/A N/A14 d 27 d (LT[α]R-Ig) 55 d (CTLA4-Ig) After 100 d or more (LT[β]R-Ig and CTLA4-Ig)N/A > 300 mg/dL- after 2 consecutive days
Streptozotocin
(200 mg/kg)
Jung et al[26]300 IEQStreptozotocin (180 mg/kg)N/A1 dROS-A - 53 d MR1 - 82 d ROS-A+MR1 - > 160 d> 200 mg/dL - 2 consecutive measurements on the same week
Påhlman et al[27]500-600AlloxanN/AN/ACsA - 16 dN/A
IntravenousAR-C117977, 10 mg/kg - > 100 d AR-C117977, 30 mg/kg - 29, 33 d
Wang et al[28]400Streptozotocin (200 mg/kg)3-4 d3 dB7-H4 - approximately 60 d> 250 mg/dL after primary graft success
Ad-LacZ - approximately > 20 d
Chen et al[29]3000 IEQStreptozotocin dissolved in saline (50 mg/kg)N/A1 wk13 wk (SGA - microencapsulated) 7 wk (ABa- microencapsulated) 5 wk (APA- microencapsulated)N/A
Giraud et al[30]1400 IEQStreptozotocin (250 mg/kg), intraperitonealN/AN/ASCOT + PEG - Solution 20 kDa ³10 g/L - 20 d, CMRL-1066 + 1% BSA - Solution 17.5 ± 1 d, UW-Solution - 17.2 ± 0.4 d, SCOT without PEG -14 ± 0.9 d HBSS + 0.5% BSA - Solution - 14 ± 0.7 d> 200 mg/dL - 2 consecutive measurements
Qi et al[31]1940 (± 39)Streptozotocin (55 mg/kg)7 dN/AN/AN/A
Potiron et al[32]800-1000Streptozotocin (180 mg/kg)1 wk4 d24.3 ± 9.7 d250 mg/dL on 2 successive measurements
Jahr et al[33]700-900Streptozotocin (55 mg/kg)7-10 dRight after transplantation1 wk> 300 mg/dL after 8.9 ± 0.7 d
Figure 1
Figure 1 Quantitative and comparative analysis of the different donor strains (A) and recipient strains (B).
DISCUSSION

Islet transplantation has the potential to provide an adequate supply of insulin to the transplanted patient and provide a solution to the problem of islet donor shortage[6].

The first successful islet transplantation for the surgical treatment of diabetes occurred in 1990 by Shapiro et al[7]. Insulin independence was achieved in a patient with type 1 diabetes at one month post-transplant. However, many technical difficulties were found that needed to be overcome to continue the development of this technique and reproduce this experiment. In the decade between 1991 and 2000, 450 islet transplantation attempts were made in type 1 diabetic patients, with a success rate of only 8%. Fifty percent of successful cases were reported when patients had become diabetic because they were undergoing a pancreatectomy.

Then, in 1999/2000, Shapiro et al[7] successfully achieved insulin independence in 7 diabetic patients by performing experiments based on the modified Edmonton protocol[2,7].

Islet transplantation has increasingly been shown to reduce morbidity 20-fold compared to pancreas transplant because it is surgically less invasive[8].

In the present study, we reviewed studies consisting of rodents similar in age and weight undergoing allograft transplantation. Strains of mice aged 6-12 wk and weighing 200 g to 350 g were used. According to these studies (Table 3), C57BL/6 (B6) and Balb/c were the most commonly used strains in allograft experiments as both islet donors (23.8% and 16.67%) and as islet recipients (28.57% and 16.67%). However, no significant difference was observed in the results obtained using other strains.

The efficacy of combined immunosuppressive drug therapy on islet transplantation in rodents has been widely studied. Among these studies, Fotiadis et al[9] tested the effects of cyclosporine A (CsA) given in combination with mycophenolate mofetil (MMF) and found that the survival rate increased significantly compared to the isolated use of CsA and MMF; this observation was presumably due to its lower toxicity in the combined regimen.

Nishimura et al[11] conducted studies with tacrolimus and demonstrated a suppression in vascular endothelial growth factors, protein kinase 14 activated by mythogen, tissue factor F, specific cyclin D1 for G1/S cell division and protein kinase 4. Thus, they concluded that the drug inhibits pancreatic islet revascularization. However, hypoxia-inducible factor 1 alpha (HIF1A) was not observed. Thus, there was a minor engraftment of islets and subsequent degeneration. Furthermore, no differences were observed in gene expression between the control group and the group receiving tacrolimus.

Wee et al[14] studied the effects of tautomycetin and concluded that it does not affect the viability of the islets and spleen, but it is capable of inhibiting the proliferation of T cells. When tautomycetin was combined with subtherapeutic doses of CsA, it led to increased survival of islets. A dose of CsA of 15 mg/kg prolonged the survival of islets the longest. Thus, the mixture of tautomycetin with CsA or other calcineurin inhibitors increased islet survival.

Merani et al[10] demonstrated that inhibition of PKC using the new drug AEB -071 slowed the rejection of islet allografts in rodents. Furthermore, addition of CsA therapy with 5 mg/kg AEB prevented graft rejection in 80% of the rats transplanted by immunosuppressive action of the complement system and had no toxic effects.

Watanabe et al[16] conducted studies with DHMEQ, an inhibitor of NF-κB, and concluded that the proinflammatory responses activated by HMGB1 were reduced. Moreover, the immunosuppression allows allograft acceptance even in cases where only a few islets were transplanted.

Xekouki et al[18] analyzed the effects of CsA and MMF. Their results suggest a beneficial effect of MMF in maintaining the architecture of the islets without prominent side effects in other organs, such as the kidneys or liver.

Baker et al[19] studied CXCR3 gene deletion and αIP-10 antibody therapy and concluded that they modulate post-transplant lymphocytic infiltration into the graft and contribute to prolonging allograft survival.

Fan et al[25] concluded that the simultaneous blockade of LIGHT and CD28 prolongs graft survival because of a synergistic effect; the presence of T-regulatory cell activity develops donor-specific immunological tolerance. Moreover, prevention of allograft rejection and induction of donor-specific tolerance in lymphocyte-sufficient recipients can be achieved by local cotransplantation of the allografts with regulatory T cells.

Jung et al[26] concluded that the combination of Ros A and MR1 in a murine allogeneic islet transplantation model prolonged graft survival compared to the MR1-alone treatment group.

Påhlman et al[27] evaluated the immunosuppressive limitations of AR-C117977, an immunosuppressant drug that maintains long-term graft survival and induces operational tolerance, and concluded that AR-C117977 combined with CsA resulted in significant prolongation of graft survival compared with AR-C117977 or CsA therapy alone. Furthermore, CsA therapy alone did not prevent acute rejection.

Wang et al[28] studied local expression of B7-H4 and concluded that it prolongs islet allograft survival in vivo.

Studies investigating immunosuppressant drugs and their toxic effects on islets in vivo are still in development. The most utilized immunosuppressants were CsA, MMF and CTLA4 Ig, as shown in Table 4. The concomitant use of glucocorticoids was associated with high rejection rates and is not recommended. Their immunosuppressive and toxic effects have not been rigorously tested, and studies are still underway.

In relation to the different locations for transplantation studied according to the table, the kidney capsule was the most frequently used site for transplantation. Second was the portal vein in the liver[14]. The spleen[9,19], intraperitoneal site[32], bone marrow (tibia)[13], dorsal skin fold-intra-abdominal[30] nonmetallic chamber[11] or subcutaneous[22] was used in a small percentage of studies.

In our review of the studies, it was found that the highest survival rate was obtained by Wee et al[14], who used the portal vein (liver) as the site of allograft transplantation and sacrificed the mice at 100 d postoperative. Melzi et al[23], Watanabe et al[16] and Merani et al[10] obtained a survival rate of 100 d, where the site of engraftment was the kidney capsule.

It is important to note that there was no standard level of hyperglycemia that the mice must present to be recipients of islet transplantation. A range of blood glucose levels from 180 mg/dL to 500 mg/dL was observed, as shown in Table 7.

Table 7 Analysis of the parameters of hyperglycemia.
Ref.Blood Glucose Levels: hyperglycemia (mg/dL)
Fotiadis et al[9]180 on 2 consecutive measurements
Merani et al[10]180 on 3 consecutive days
Makhlouf et al[12]Moderate diabetes: between 240 and 350
Severe diabetes: between 351 and 550
Very severe diabetes: more than 550
Wee et al[14]200
Plesner et al[15]≥ 360
Watanabe et al[16]200 on 2 consecutive days
Gysemans et al[17]200 on 2 consecutive days
Xekouki et al[18]300
Baker et al[19]300 on 3 consecutive days
Li et al[20]350 to 500 after 2 consecutive measurements
Melzi et al[23]250 on 2 consecutive measurements
Fan et al[25]300 on 2 consecutive measurements
Jung et al[26]300 on 2 consecutive days
Påhlman et al[27]288 on 2 consecutive measurements
Wang et al[28]300 after 2 consecutive days
Chen et al[29]302.4 on more than 3 consecutive days
Giraud et al[30]350 after 2 consecutive days
Qi et al[31]450
Potiron et al[32]> 200

The articles have many independent variables that influence the study results, such as species of rodent, immunosuppressant drugs and dosages, criteria for diabetes and allograft site. Thus, more research is needed to develop the ideal allograft model of islet transplantation.

CONCLUSION

Based on the analyzed studies, we can infer that islet allotransplantation in rodents is promising and continues to develop. The survival rates of allografts have increased with the discovery of new immunosuppressive drugs and the use of different graft sites. These advancements have the potential to overcome the scarcity of islet donors and improve the treatment of type 1 diabetes.

Footnotes

P- Reviewer: Oh SH S- Editor: Qi Y L- Editor: A E- Editor: Du P

References
1.  Delfino VDA, Mocelin AJ. Transplante de Pâncreas e de Ilhotas Pancreáticas: Visão de Nefrologista. Arq Bras Endocrinol Metab. 2002;46:177-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
2.  Chaves G Incidência de diabetes tipo 1 aumenta 3% ao ano. Minas Gerais: Saúde Plena; 2013; [updated 2013 July 21; cited 2013 July 22] Available from: http://sites.uai.com.br/app/noticia/saudeplena/noticias/2013/07/22/noticia_saudeplena,144088/incidencia-de-diabetes-tipo-1-aumenta-3-ao-ano.shtml.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Sociedade Brasileira de Diabetes Internet. São Paulo: Sociedade Brasileira de Diabetes; 2012 [cited 2012 April 18].  Available from: http://www.diabetes.org.br/sala-de-noticias/2116-sao-12-milhoes-de-diabeticos-no-brasil.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Merani S, Shapiro AM. Current status of pancreatic islet transplantation. Clin Sci (Lond). 2006;110:611-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 105]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
5.  United States of America National Institute of Diabetes and Digestive and Kidney Diseases. Pancreatic Islet Transplantation. USA: NIH Publication 07-4693 2007; .  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Eliaschewitz FG, Franco DR, Mares-Guia TR, Noronha IL, Labriola L, Sogayar MC. [Islet transplantation as a clinical tool: present state and future perspectives]. Arq Bras Endocrinol Metabol. 2009;53:15-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
7.  Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343:230-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3996]  [Cited by in F6Publishing: 3720]  [Article Influence: 155.0]  [Reference Citation Analysis (0)]
8.  Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, Lakey JR, Shapiro AM. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54:2060-2069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1275]  [Cited by in F6Publishing: 1173]  [Article Influence: 61.7]  [Reference Citation Analysis (0)]
9.  Fotiadis C, Xekouki P, Papalois AE, Antonakis PT, Sfiniadakis I, Flogeras D, Karampela E, Zografos G. Effects of mycophenolate mofetil vs cyclosporine administration on graft survival and function after islet allotransplantation in diabetic rats. World J Gastroenterol. 2005;11:2733-2738.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Merani S, Pawlick RL, Edgar RL, Toso C, Emamaullee J, Anderson CC, Shapiro AM. Protein kinase C inhibitor, AEB-071, acts complementarily with cyclosporine to prevent islet rejection in rats. Transplantation. 2009;87:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
11.  Nishimura R, Nishioka S, Fujisawa I, Shiku H, Shimada M, Sekiguchi S, Fujimori K, Ushiyama A, Matsue T, Ohuchi N. Tacrolimus inhibits the revascularization of isolated pancreatic islets. PLoS One. 2013;8:e56799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
12.  Makhlouf L, Duvivier-Kali VF, Bonner-Weir S, Dieperink H, Weir GC, Sayegh MH. Importance of hyperglycemia on the primary function of allogeneic islet transplants. Transplantation. 2003;76:657-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
13.  Salazar-Bañuelos A, Wright JR, Sigalet D, Benítez-Bribiesca L. Pancreatic islet transplantation into the bone marrow of the rat. Am J Surg. 2008;195:674-678; discussion 678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
14.  Wee YM, Choi MY, Kang CH, Kim YH, Kim JH, Lee SK, Yu SY, Kim SC, Han DJ. The synergistic effect of Tautomycetin on Cyclosporine A-mediated immunosuppression in a rodent islet allograft model. Mol Med. 2005;16:298-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
15.  Plesner A, Liston P, Tan R, Korneluk RG, Verchere CB. The X-linked inhibitor of apoptosis protein enhances survival of murine islet allografts. Diabetes. 2005;54:2533-2540.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Watanabe M, Yamashita K, Kamachi H, Kuraya D, Koshizuka Y, Shibasaki S, Asahi Y, Ono H, Emoto S, Ogura M. Efficacy of DHMEQ, a NF-κB inhibitor, in islet transplantation: II. Induction DHMEQ treatment ameliorates subsequent alloimmune responses and permits long-term islet allograft acceptance. Transplantation. 2013;96:454-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
17.  Gysemans CA, Waer M, Valckx D, Laureys JM, Mihkalsky D, Bouillon R, Mathieu C. Early graft failure of xenogeneic islets in NOD mice is accompanied by high levels of interleukin-1 and low levels of transforming growth factor-beta mRNA in the grafts. Diabetes. 2000;49:1992-1997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
18.  Xekouki P, Papalois A, Fotiadis C, Sfiniadakis J, Karampela E, Papadopoulou A, Grigoriou T, Sechas MN. In vivo test of two low doses of mycophenolate mofetil in an experimental model of islet allotransplantation. Transplant Proc. 2002;34:1446-1448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
19.  Baker MS, Chen X, Rotramel AR, Nelson JJ, Lu B, Gerard C, Kanwar Y, Kaufman DB. Genetic deletion of chemokine receptor CXCR3 or antibody blockade of its ligand IP-10 modulates posttransplantation graft-site lymphocytic infiltrates and prolongs functional graft survival in pancreatic islet allograft recipients. Surgery. 2003;134:126-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
20.  Li X, Chen H, Epstein PN. Metallothionein protects islets from hypoxia and extends islet graft survival by scavenging most kinds of reactive oxygen species. J Biol Chem. 2004;279:765-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 121]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
21.  Vieiro M, Ceballos C, Barrionuevo P, Hyon SH, Isturiz M, Argibay P. Inhibition of allospecific response in pancreatic islet transplantation: the glycan approach. Transplant Proc. 2005;37:243-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
22.  Neuzillet Y, Giraud S, Lagorce L, Eugene M, Debre P, Richard F, Barrou B. Effects of the molecular weight of peg molecules (8, 20 and 35 KDA) on cell function and allograft survival prolongation in pancreatic islets transplantation. Transplant Proc. 2006;38:2354-2355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
23.  Melzi R, Battaglia M, Draghici E, Bonifacio E, Piemonti L. Relevance of hyperglycemia on the timing of functional loss of allogeneic islet transplants: implication for mouse model. Transplantation. 2007;83:167-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Fiorina P, Jurewicz M, Tanaka K, Behazin N, Augello A, Vergani A, von Andrian UH, Smith NR, Sayegh MH, Abdi R. Characterization of donor dendritic cells and enhancement of dendritic cell efflux with CC-chemokine ligand 21: a novel strategy to prolong islet allograft survival. Diabetes. 2007;56:912-920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
25.  Fan K, Wang H, Wei H, Zhou Q, Kou G, Hou S, Qian W, Dai J, Li B, Zhang Y. Blockade of LIGHT/HVEM and B7/CD28 signaling facilitates long-term islet graft survival with development of allospecific tolerance. Transplantation. 2007;84:746-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
26.  Jung da Y, Kim EY, Joo SY, Park JB, Moon C, Kim SH, Sim EY, Joh JW, Kwon CH, Kwon GY. Prolonged survival of islet allografts in mice treated with rosmarinic acid and anti-CD154 antibody. Exp Mol Med. 2008;40:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
27.  Påhlman C, Malm H, Qi Z, Veress B, Ferguson D, Bundick R, Murray C, Donald D, Ekberg H. Operational tolerance in nonvascularized transplant models induced by AR-C117977, a monocarboxylate transporter inhibitor. Transplantation. 2008;86:1135-1138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
28.  Wang X, Hao J, Metzger DL, Mui A, Ao Z, Verchere CB, Chen L, Ou D, Warnock GL. Local expression of B7-H4 by recombinant adenovirus transduction in mouse islets prolongs allograft survival. Transplantation. 2009;87:482-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
29.  Chen X, Shao W, Chen JB, Zhang L, Matthias C, Shan SG, Qi ZQ. Allotransplantation of sulphate glucomannan-alginate barium (SGA)-microencapsulated rat islets for the treatment of diabetes mellitus. Immunol Invest. 2009;38:561-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  Giraud S, Bon D, Neuzillet Y, Thuillier R, Eugene M, Hauet T, Barrou B. Concentration and chain length of polyethylene glycol in islet isolation solution: evaluation in a pancreatic islet transplantation model. Cell Transplant. 2012;21:2079-2088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
31.  Qi Z, Yamamoto C, Imori N, Kinukawa A, Yang KC, Yanai G, Ikenoue E, Shen Y, Shirouzu Y, Hiura A. Immunoisolation effect of polyvinyl alcohol (PVA) macroencapsulated islets in type 1 diabetes therapy. Cell Transplant. 2012;21:525-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
32.  Potiron N, Chagneau C, Boeffard F, Soulillou JP, Anegon I, Le Mauff B. Adenovirus-mediated CTLA4Ig or CD40Ig gene transfer delays pancreatic islet rejection in a rat-to-mouse xenotransplantation model after systemic but not local expression. Cell Transplant. 2005;14:263-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
33.  Jahr H, Hussmann B, Eckhardt T, Bretzel RG. Successful single donor islet allotransplantation in the streptozotocin diabetes rat model. Cell Transplant. 2002;11:513-518.  [PubMed]  [DOI]  [Cited in This Article: ]