Open access peer-reviewed chapter

Pathology of Intestinal Transplantation: Rejection and a Case of Tolerance

Written By

Tatsuaki Tsuruyama

Submitted: 17 July 2020 Reviewed: 06 October 2020 Published: 10 November 2020

DOI: 10.5772/intechopen.94361

From the Edited Volume

Organ Donation and Transplantation

Edited by Vassil Mihaylov

Chapter metrics overview

493 Chapter Downloads

View Full Metrics

Abstract

Small bowel transplants are less common than other organ transplants. Histological criteria for rejection of the transplanted small intestine were proposed at the 8th International Symposium on Small Intestinal Transplantation 2003-2004. The Banff Conference on Transplant Disease Pathology, an international conference on the rejection of small bowel transplants, was held in 2019, and unifying diagnostic criteria were discussed (https://banfffoundation.org/pittsburgh-2019/). These histological criteria are expected to be standardized in the near future. This review outlines new findings such as apoptosis and apoptotic-body phagocytic findings in the lamina propria and behavior of natural killer T (NKT) cells, in addition to previously known crypt Fas-related apoptosis in acute cellular rejection. Furthermore, we review the case of a recipient who has shown no rejection for 5 years after transplantation. In the transplanted small intestine of this patient, the lymphocytes were replaced by those of another male patient.

Keywords

  • intestinal transplantation
  • histology
  • rejection
  • natural killer T cells
  • apoptosis
  • tolerance

1. Introduction

1.1 Current status of small bowel transplantation

Small bowel transplantation (SBT) is one of the standard treatments for patients who are unable to consume a regular diet and have complications from the irreversible requirement of parenteral nutrition [1]. Hirschsprung’s disease [2, 3] and Crohn’s disease [4] patients are two examples. Recent effective immunosuppressive drugs, well-controlled postoperative care, and advances in diagnostic techniques have significantly improved the outcome of SBT [5]. Immunosuppressants such as mycophenolate mofetil, tacrolimus, and steroids, are routinely used for long-term management after transplantation [6, 7].

Acute cellular rejection (ACR) is a major cause of impaired colonization by the transplanted small intestine, and it frequently accompanies chronic and irreversible changes such as ulcers and lamina propria fibrosis. ACR has remained a risk factor that impedes functional recovery of the intestinal graft [1, 8, 9]. On the other hand, pathologists frequently encounter various pathologies of the intestinal allograft [10, 11, 12]. For example, mechanical failure of the graft due to operation during surgery may occur during the early phase after transplantation. Cytomegalovirus (CMV) enteritis and Epstein–Barr virus -related enteritis are severe side effects and post-transplantation lymphoproliferative disorders/diseases [13, 14, 15]. It is often difficult to make a differential diagnosis of the ACR findings. However, histologic diagnosis is critical for the selection of immunosuppressants and their respective doses. Tacrolimus, cyclosporin, and steroids are commonly prescribed in the early stages of rejection [16]. If an excessive dose is administered, the occurrences of CMV enteritis and EBV enteritis become inevitable.

Among the various histological features, crypt epithelial cell apoptosis has been evaluated as a highly reproducible finding. However, other histological findings have been proposed at different institutions. We have also previously suggested other findings as indicators of ACR [17, 18, 19].

Advertisement

2. Diagnostic criteria for ACR

2.1 Crypt apoptosis

Crypt apoptosis is considered a unique feature of ACR in SBT. The crypt is an architectural element that is located at the base of the villous epithelium and serves as the source of mucosal cells. Paneth cells, stem cells, and reserve stem cells are included in the crypt. Enterocytes are differentiated from reserve stem cells in the crypt and migrate to the tips of villi through the transit amplifying zone [20]. The kinetics of differentiation and loss of enterocytes contribute to the maintenance of quick renewal for mucosal homeostasis. The supply of enterocytes becomes interrupted by apoptosis in the crypt, and the shortening of villi becomes unavoidable. When ulceration occurs, the lesion is susceptible to infectious enteritis such as CMV- and EBV-related enteritis, for a significant period of time [21, 22].

Pathologically, the diagnosis of small bowel transplant rejection is based on the appearance of 6 or more apoptotic lesions per ten crypts [3, 4] (Table 1). The detection of crypt apoptosis is commonly used because of its high reproducibility. Nevertheless, discussions about the number of lesions per crypt were held at the Banff Conference 2019. In the case we experienced, if more than six apoptotic cells were detected in the crypts, subsequent ulceration is inevitable, and infection from the ulcer site might occur. Therefore, we considered that immunosuppressants should be administered when apoptotic cells were observed in the crypt [18].

Histologic grade
IndeterminateCrypt apoptosis and related findingsLymphocytic apoptosis in the lamina propria [18]
Up to 6 apoptotic bodies per 10 cryptsNone
Mild>6 apoptotic bodies per 10 crypts
Confluent apoptosis
Isloated apoptotic bodies in the lamina propriaPhagocytosis of apoptotic bodies by macrophages [18]
ModerateIncreased inflammation, epithelial injuryA few apoptotic body cluster in the lamina propriaAggregation of macrophages [18]
Severe/exfoliativeMucosal ulcerationApoptotic bodies aggregate in the lamina propriaGranuloma consisting of macrophages

Table 1.

Histological criteria for ACR of the intestinal allograft [10]. The findings under the lymphocyte and macrophage categories refer to our previous study [18].

Previous apoptosis findings have shown that the cells are eosinophilic with an intensely stained nucleus [17, 18] (Figure 1). Cells with lobulated nuclei, such as neutrophils and apoptotic cells, can be confused morphologically; therefore, careful observation is necessary. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining is one method to avoid this confusion. This staining procedure involves an enzyme-mediated reaction. First, the fragmented DNA is labeled with biotin containing terminal deoxynucleotidyl transferase. The labeled DNA then reacts with streptavidin for staining. Both labels with 3,3′-diaminobenzidine (DAB) and fluorescein isothiocyanate (FITC) are available for visualization of the apoptotic body [18].

Figure 1.

Histology of ACR of the intestinal allograft. The onset of ACR. Eosinophil infiltrates are observed in the ulcerated mucosa (left, 100×). Apoptotic bodies are observed in the crypt (indicated by arrows, middle, 200×; right, 100×, TUNEL-stained with 3,3’-diaminobenzidine).

As apoptosis progresses, fragmented cell debris (apoptotic bodies) are observed in or around the crypt. Increasing the dose of the immunosuppressive drug suppresses the progression of apoptosis. Therefore, quick detection of apoptosis is critical for effective immunosuppression therapy [18, 19].

The factors that cause such apoptotic responses in the crypt and lamina propria are poorly understood. It is possible that cytotoxic T lymphocytes (CTLs) can directly attack the crypt of the graft. However, it is not always histologically evident that CTLs directly infiltrate near the crypt and remain near this area. There is also a noteworthy research report suggesting that CD8-positive CTLs are not always involved in ACR [23]. At the basic research level, rejection of the apoptosis-inducing factors perforin and granzyme B released from CTLs has been reported [24]. Therefore, the destruction of the mucosal immune system by local increases in complement and inflammatory cytokines is thought to be the cause of apoptosis.

2.2 Immunohistochemical monitoring

In addition to the crypt apoptosis, apoptotic lymphocytes are identified by systematic immunostaining of lymphocyte surface antigens: T cell surface antigens CD3, CD4, and CD8; B cell surface antigens CD20 and CD79a; natural killer cell surface antigen CD56; and activated lymphocytes Fas and its ligand (FasL) [25]. FasL, also known as CD95L, is a surface antigen of activated cytotoxic T cells and NK cells are observed at the onset of rejection [18] (Figure 2, upper panels).

Figure 2.

FasL immunostaining of the intestinal allograft. FasL-positive lymphocytes in the lamina propria (upper left, 200×) and Peyer’s patch (upper right, 400×) are shown. FasL-stained apoptotic bodies (lower left, 400×). Apoptotic TCRVα24 stained cells (lower right, 400×). TCRVα24 and FasL were visualized with DAB (3,3’-diaminobenzidine).

Apoptotic bodies are also been observed in the lamina propria and Peyer’s patch (PP) distant from the crypt, and the macrophages that phagocytose them often aggregate to present granuloma-like findings. Notably, these bodies are stained with FasL and Fas, suggesting that the apoptosis relates to the FasL-Fas interactive reaction (Figure 2, lower panels). This result was first reported in our previous study [18].

Advertisement

3. Endoscopic examination and Peyer’s patch response

Endoscopically, elevation of the small intestinal mucosa may be recognized and biopsied when clinical rejection is suspected. Since this elevation is observed in patients who are not receiving oral nutrition, the change may not be the result of irritation from the lumen of the small intestine and more likely due to the reaction of the Peyer’s patches (PPs) to a load of patient cells on the graft mucosal immune system. In our cases, the biopsied Peyer’s patches were injured at the onset of ACR (Figures 3A and B). Therefore, PP is one of the targets of ACR or other types of rejection (Figure 3C). Notably, B cells increased in number in the disintegrated PPs (Figure 3). As described later, IL-5 was increased in the intestinal allograft [17], which may promote the transient B cell growth in PP.

Figure 3.

Histology of a PP in an intestinal allograft. (A, B) A hyperplastic Peyer’s patch stained with CD79a antibody before ACR (A) and at the onset of ACR (B). (C) CD8 staining of PP after 42 h at the onset of rejection. Many CD8+ CTLs infiltrate in PP. CD79 and CD8 were visualized by DAB. The photo magnitude is 100×.

Advertisement

4. Cases at Kyoto University Hospital

Here we review cases of SBT at Kyoto University Hospital [17, 18, 21, 22]. SBT was performed owing to intestinal malrotation and Hirschsprung’s disease-related effects (Figure 4).

Figure 4.

Immunofluorescent staining of natural killer T cells in the intestinal allograft. Immunostaining of an intestinal allograft. Green signal, FITC and red signal, phycoerythrin [PE]. Nuclei are stained with DAPI (blue). Brown signal was visualized with DAB. (A) TCRVα24 (200×) and (B) TCRβ11 (200×). (C, D) TCRVα24 (green) and IL-4 (red) (IL-4 positive iNKT is indicated by an arrow). The observation magnification is 200× in both cases. (E, F) TCRVα24 (red) and TUNEL (green). (E) TUNEL+ (apoptotic) TCRVα24 + iNKT cells are observed at the onset of ACR (100×) and (F) 48 h after the onset of ACR (100×). Doubly stained cells were increased 48 h after the onset of ACR. (G) CD1d+ dendritic cells. CD1d and CD11c were stained green and red, respectively. (H) TCRVα24 stained iNKT cells (red) and CD1d stained dendritic cells (green). (I) FasL+ (green) TCRVα24+ (red) iNKT cells. The observation magnification is 400 x in (G)-(I).”

Jejunal or ileal grafts were monitored histologically. When fever, increased intestinal juice, abdominal pain, or C-reactive protein (CRP) elevation in peripheral blood (>0.5 mg/10−1 L) was observed, an endoscopic examination was performed. In particular, for the first 1 to 2 weeks after surgery, the examination was performed every other day, and a histological examination was also performed. Once the condition of the patient became stable, a histological examination was performed approximately once a week, and the state of the intestinal graft was monitored continuously for up to 2 months in the hospital. The patient received immunosuppressive therapy in combination with tacrolimus (trough concentration: 20 ng/mL) and methylprednisolone (30 mg/kg/day, 1 to 3 times). In the biopsy examination, diagnosis by hematoxylin and eosin staining and findings specific to rejection within 6 h were confirmed by immunostaining of frozen sections. For histological diagnosis, we stained the apoptosis-related proteins such as FasL and surface antigens of B cells, T cells, and NK cells in each case. Steroid pulse therapy was conducted following detecting the immunological activation with the appearance of FasL-positive T/ NKT cells and apoptotic bodies in the lamina propria. The treatment substantially prevented the progression of the crypt apoptosis [17, 18, 21, 22].

Advertisement

5. Cytokine production in the intestinal allograft

5.1 NKT cells and cytokines

NKT cells are resident in the large bowel and increase in number in the colorectal cancer tissue [26]. The NKT cells have a limited T cell repertoire, and the restricted types are called invariant types of NKT (iNKT) cells. During the onset of intestinal rejection, the α chain 24 (TCRVα24) and β chain 11 (TCRVβ11) on iNKT cells are positively stained (Figure 4A and B) [17]. iNKT cells are mainly involved in innate immunity against glycolipids with the assistance of CD1d + dendritic cells [27]. Since iNKT cells are not identified in the small intestine of healthy donors before transplantation, this finding to be an indicator of ACR [17, 28].

Th1 cytokines, such as interferon-gamma (IFN-γ), generally act on the differentiation of CTLs, which promote rejection, while Th2 cytokines may suppress ACR of SBT. TCRVα24 (+) invariant NKT (iNKT) cells are positive for interleukin 4 (IL-4) in allografts of the intestine during rejection (Figure 4C and D) [8]. The apoptosis of iNKT cells are observed at the onset of rejection (Figure 4E and F), indicating that a part of apoptotic cells in the lamina propria are iNKT cells (Figure 2, lower right). CD1d+ dendritic cells are detected during the rejection process at the same time that the rejection progressed (Figure 4G and H). The involvement of iNKT cells in the rejection reaction has been discussed previously, and there is also an experimental report regarding their involvement in tolerance [29, 30]. However, the involvement of iNKT cells in rejection has not yet become apparent [31]. Furthermore, the mechanism by which the expression of IL-4 is directly involved in mucosal immune regulation remains unclear. However, IL-4 may suppress the action of CTLs that cause rejection. On the other hand, iNKT cells expressed FasL, indicating that they are activated in ACR (Figure 4I).

In addition, increased IL-5 production is also observed at the onset of rejection. IL-5 promotes eosinophil differentiation and chemotaxis [32]. This increase in production may explain the large number of eosinophils infiltrating the mucosa at the time of rejection [17]. Conventional T cells and iNKT cells may secrete IL-5 [17]. The role of eosinophils in rejection has often been debated [33] and there is a discussion on whether eosinophils may be the target of rejection therapy [34]. An increase in the rejection of eosinophils has also been reported in the transplanted liver [35]. In the small intestine, the presence of the mucosal immune system may further complicate the graft’s immunological environment. Increased eosinophils, however, are histologically detectable and may provide useful information for the diagnosis of rejection, even in small bowel transplant grafts [5]. As a result of an imbalance in mucosal immunity, excess production of IL-4 and IL-5 may damage the mucosal epithelium. The administration of immunosuppressive drugs acts on iNKT cells in addition to cytotoxic T cells. Therefore, the distribution of immunocompetent lymphocytes in the mucosa is disturbed, and the treatment protocol should be developed further.

Advertisement

6. Histological tolerance of the intestinal allograft

Finally, we reviewed a case of histological tolerance reported [25]. This case involves a transplant in a 4-year-old male patient who had short bowel syndrome and previously underwent a living small bowel transplant from his mother who was in her twenties. The patient underwent a small intestinal biopsy 2–3 times per week for one month. Immunological analysis was performed using CD3, CD4, CD8, CD20, CD56, CD79a, perforin, granzyme B, FasL, Fas, and TUNEL staining. No severe rejection with an increase in FasL-positive T cells was detected. The maximum level of CRP, an inflammation marker, was 1.0 (mg/10−1 L) at POD67. In situ hybridization was performed using a Y-chromosome probe to evaluate rejection or tolerance for evaluation of the immunologic stability of the graft and chimerization [36], which comprises multiplex staining with a CD3 fluorescent substance, for monitoring allografts. Figure 5 shows photographs of the graft 5 years after transplantation. A part of native T lymphocytes were replaced with Y-chromosome positive T lymphocytes from a male patient. This patient has been living for longer than ten years without any clinical symptoms, such as rejection, and is likely one of the first cases of operational tolerance.

Figure 5.

Combined in situ hybridization of lymphocytes with the Y-chromosome probe (red: PE) and CD3-lymphocytic immunohistochemistry (green: FITC). The photos show the double-stained T cells carrying the Y-chromosomal investigation, indicating the male-donor derived lymphocytes in the female-derived intestinal allograft. Left (100×) and right (400×). The nuclei were stained red, indicating Y-chromosome positivity.

Advertisement

7. Conclusion

Early diagnosis of rejection of the transplanted small intestine is essential to facilitate the initiation of therapy that interferes with rejection progression. In addition to crypt apoptosis, apoptotic bodies in the lamina propria is considered useful for diagnosis. Furthermore, iNKT cell infiltration was another characteristic finding. Since histologic features of ACR have been studied extensively. Of note in future diagnoses are the issues of humoral and chronic rejection.

Advertisement

Appendices and nomenclature

ACRacute cellular rejection
CMVcytomegalovirus
CRPC-reactive protein
CTLcytotoxic T lymphocyte
EBVEpstein–Barr virus
FasLFas ligand
FITCfluorescein isothiocyanate
iNKT cellsinvariant natural killer T cells
IL-4interleukin 4
IL-5interleukin 5
NKT cellsnatural killer T cells
SBTsmall bowel transplantation
TUNELterminal deoxynucleotidyl transferase dUTP nick end labeling

References

  1. 1. Pirenne J, Hoffman I, Miserez M, Coosemans W, Aerts R, Monbaliu D, et al. Selection criteria and outcome of patients referred to intestinal transplantation: an European center experience. Transplant Proc. 2006;38(6):1671-1672. Epub 2006/08/16. S0041-1345(06)00553-7 [pii]. doi: 10.1016/j.transproceed.2006.05.063. PubMed PMID: 16908242.
  2. 2. Neuvonen MI, Kyrklund K, Lindahl HG, Koivusalo AI, Rintala RJ, Pakarinen MP. A population-based, complete follow-up of 146 consecutive patients after transanal mucosectomy for Hirschsprung disease. Journal of Pediatric Surgery. 2015;50(10):1653-1658. Epub 2015/03/19. PubMed PMID: 25783387. DOI: 10.1016/j.jpedsurg.2015.02.006
  3. 3. Masetti M, Rodriguez MM, Thompson JF, Pinna AD, Kato T, Romaguera RL, et al. Multivisceral transplantation for megacystis microcolon intestinal hypoperistalsis syndrome. Transplantation. 1999;68(2):228-232 Epub 1999/08/10. PubMed PMID: 10440392
  4. 4. Gerlach UA, Vrakas G, Reddy S, Baumgart DC, Neuhaus P, Friend PJ, et al. Chronic intestinal failure after Crohn disease: When to perform transplantation. JAMA Surgery. 2014;149(10):1060-1066. DOI: 10.1001/jamasurg.2014.1072 PubMed PMID: 25162284
  5. 5. Pinna AD, Weppler D, Nery J, Ruiz P, Kato T, Khan F, et al. Intestinal transplantation at the University of Miami-five years of experience. Transplant Proc. 2000;32(6):1226-1227 Epub 2000/09/21. doi: S0041134500011994 [pii]. PubMed PMID: 10995922
  6. 6. Tzakis AG, Kato T, Nishida S, Levi DM, Tryphonopoulos P, Madariaga JR, et al. Alemtuzumab (Campath-1H) combined with tacrolimus in intestinal and multivisceral transplantation. Transplantation. 2003;75(9):1512-1517 Epub 2003/06/07. doi: 10.1097/01.TP.0000060250.50591.39. PubMed PMID: 12792506
  7. 7. Tzakis AG, Nery JR, Thompson J, Webb MG, Khan FA, Khan RT, et al. New immunosuppressive regimens in clinical intestinal transplantation. Transplant Proc. 1997;29(1-2):683-685 Epub 1997/02/01. PubMed PMID: 9123479
  8. 8. Reyes J. Intestinal transplantation for children with short bowel syndrome. Seminars in Pediatric Surgery. 2001;10(2):99-104 Epub 2001/05/01. doi: S105585860100021X [pii]. PubMed PMID: 11329611
  9. 9. Avitzur Y, Grant D. Intestine transplantation in children: update 2010. Pediatr Clin North Am. 2010;57(2):415-431, table of contents. Epub 2010/04/08. doi: S0031-3955(10)00020-9 [pii]. 10.1016/j.pcl.2010.01.019. PubMed PMID: 20371045
  10. 10. Wu T, Abu-Elmagd K, Bond G, Nalesnik MA, Randhawa P, Demetris AJ. A schema for histologic grading of small intestine allograft acute rejection. Transplantation. 2003;75(8):1241-1248. Epub 2003/04/30. DOI: 10.1097/01.TP.0000062840.49159.2F PubMed PMID: 12717210
  11. 11. Ruiz P, Takahashi H, Delacruz V, Island E, Selvaggi G, Nishida S, et al. International grading scheme for acute cellular rejection in small-bowel transplantation: single-center experience. Transplant Proc. 2010;42(1):47-53 Epub 2010/02/23. S0041-1345(09)01782-5 [pii] doi: 10.1016/j.transproceed.2009.12.026. PubMed PMID: 20172279.
  12. 12. Ruiz P, Bagni A, Brown R, Cortina G, Harpaz N, Magid MS, et al. Histological criteria for the identification of acute cellular rejection in human small bowel allografts: results of the pathology workshop at the VIII International Small Bowel Transplant Symposium. Transplant Proc. 2004;36(2):335-337. Epub 2004/03/31. DOI: 10.1016/j.transproceed.2004.01.079 S0041134504000995 [pii]. PubMed PMID: 15050150
  13. 13. Charles J, Di Domizio J, Salameire D, Bendriss-Vermare N, Aspord C, Muhammad R, et al. Characterization of circulating dendritic cells in melanoma: Role of CCR6 in plasmacytoid dendritic cell recruitment to the tumor. The Journal of Investigative Dermatology. 2010;130(6):1646-1656. DOI: 10.1038/jid.2010.24 PubMed PMID: 20220766
  14. 14. Kubal CA, Pennington C, Fridell J, Ekser B, Muhaylov P, Mangus R. Challenges with Intestine and Multivisceral Re-Transplantation: Importance of Timing of Re-Transplantation and Optimal Immunosuppression. Ann Transplant. 2018;23:98-104. Epub 2018/02/07. doi: 10.12659/aot.908052. PubMed PMID: 29402878; PubMed Central PMCID: PMCPMC6248276.
  15. 15. Koo J, Dawson DW, Dry S, French SW, Naini BV, Wang HL. Allograft biopsy findings in patients with small bowel transplantation. Clinical Transplantation. 2016;30(11):1433-1439. Epub 2016/11/03. PubMed PMID: 27582272. DOI: 10.1111/ctr.12836
  16. 16. Jiang JW, Ren ZG, Lu HF, Zhang H, Li A, Cui GY, et al. Optimal immunosuppressor induces stable gut microbiota after liver transplantation. World J Gastroenterol. 2018;24(34):3871-3883. Epub 2018/09/20. doi: 10.3748/wjg.v24.i34.3871. PubMed PMID: 30228781; PubMed Central PMCID: PMCPMC6141331.
  17. 17. Tsuruyama T, Fujimoto Y, Yonekawa Y, Miyao M, Onodera H, Uemoto S, et al. Invariant natural killer T cells infiltrate intestinal allografts undergoing acute cellular rejection. Transpl Int. 2012;25(5):537-544. Epub 2012/03/03. doi: 10.1111/j.1432-2277.2012.01450.x. PubMed PMID: 22380521.
  18. 18. Tsuruyama T, Okamoto S, Fujimoto Y, Yoshizawa A, Yoshitoshi E, Egawa H, et al. Histology of intestinal allografts: Lymphocyte apoptosis and phagocytosis of lymphocytic apoptotic bodies are diagnostic findings of acute rejection in addition to crypt apoptosis. The American Journal of Surgical Pathology. 2013;37(2):178-184. DOI: 10.1097/PAS.0b013e31826393fe PubMed PMID: 23026933
  19. 19. Tsuruyama T. Histopathology of intestinal transplant rejections. SOJ immunol. 2016;4(1):1-7
  20. 20. Baulies A, Angelis N, Li VSW. Hallmarks of intestinal stem cells. Development. 2020;147(15). Epub 2020/08/05. doi: 10.1242/dev.182675. PubMed PMID: 32747330.
  21. 21. Yoshitoshi EY, Yoshizawa A, Ogawa E, Kaneshiro M, Takada N, Okamoto S, et al. The challenge of acute rejection in intestinal transplantation. Pediatric Surgery International 2012;28(8):855-859. doi: 10.1007/s00383-012-3110-x. PubMed PMID: 22760434.
  22. 22. Fujimoto Y, Uemoto S, Inomata Y, Egawa H, Fujita S, Kawanami T, et al. Small bowel transplantation using grafts from living-related donors. Two case reports. Transpl Int. 2000;13 Suppl 1:S179-S184. Epub 2000/12/09. PubMed PMID: 11111992.
  23. 23. Krams SM, Hayashi M, Fox CK, Villanueva JC, Whitmer KJ, Burns W, et al. CD8+ cells are not necessary for allograft rejection or the induction of apoptosis in an experimental model of small intestinal transplantation. J Immunol. 1998;160(8):3673-3680. Epub 1998/04/29. PubMed PMID: 9558067.
  24. 24. McDiarmid SV, Farmer DG, Kuniyoshi JS, Robert M, Khadavi A, Shaked A, et al. Perforin and granzyme B. Cytolytic proteins up-regulated during rejection of rat small intestine allografts. Transplantation. 1995;59(5):762-766. Epub 1995/03/15. PubMed PMID: 7886805.
  25. 25. Tsuruyama T. Histological image of transplanted small intestine. Japanese Journal of Pediatric Surgery. 2013;45(7):711-715. (In Japanese)
  26. 26. Tachibana T, Onodera H, Tsuruyama T, Mori A, Nagayama S, Hiai H, et al. Increased intratumor Valpha24-positive natural killer T cells: a prognostic factor for primary colorectal carcinomas. Clin Cancer Res. 2005;11(20):7322-7327. Epub 2005/10/26. doi: 11/20/7322 [pii]. 10.1158/1078-0432.CCR-05-0877. PubMed PMID: 16243803.
  27. 27. van Dieren JM, van der Woude CJ, Kuipers EJ, Escher JC, Samsom JN, Blumberg RS, et al. Roles of CD1d-restricted NKT cells in the intestine. Inflamm Bowel Dis. 2007;13(9):1146-1152. Epub 2007/05/04. doi: 10.1002/ibd.20164. PubMed PMID: 17476670.
  28. 28. Tsuruyama T, Aini W. The Roles of Invariant NKT Cells in Bowel Immunity — Suppression of Tumor Progression and Rejection of Intestinal Transplants. 2014. doi: 10.5772/57588.
  29. 29. Chu X, Kilpatrick E, Xiao X, Liu W, Demirci G, Exley M, et al. Islet allograft tolerance in the absence of invariant natural killer T cells. Clinical Immunology 2011;141(3):268-272. doi: 10.1016/j.clim.2011.09.003. PubMed PMID: 21996456; PubMed Central PMCID: PMC3221878.
  30. 30. Brossay L, Chioda M, Burdin N, Koezuka Y, Casorati G, Dellabona P, et al. CD1d-mediated recognition of an alpha-galactosylceramide by natural killer T cells is highly conserved through mammalian evolution. Journal of Experimental Medicine. 1998;188(8):1521-1528. PubMed PMID: ISI:000076620100014.
  31. 31. Vasan S, Tsuji M. A double-edged sword: The role of NKT cells in malaria and HIV infection and immunity. Seminars in Immunology 2010;22(2):87-96. doi: 10.1016/j.smim.2009.11.001. PubMed PMID: 19962909; PubMed Central PMCID: PMC3603358.
  32. 32. Rozenberg P, Reichman H, Zab-Bar I, Itan M, Pasmanik-Chor M, Bouffi C, et al. CD300f:IL-5 cross-talk inhibits adipose tissue eosinophil homing and subsequent IL-4 production. Sci Rep. 2017;7(1):5922. Epub 2017/07/21. doi: 10.1038/s41598-017-06397-4. PubMed PMID: 28725048; PubMed Central PMCID: PMCPMC5517555.
  33. 33. Bush JW, Mohammad S, Melin-Aldana H, Kagalwalla AF, Arva NC. Eosinophilic density in graft biopsies positive for rejection and blood eosinophil count can predict development of post-transplant digestive tract eosinophilia. Pediatric Transplantation 2016;20(4):540-551. Epub 2016/02/27. doi: 10.1111/petr.12693. PubMed PMID: 26917244.
  34. 34. Kers J, Florquin S. Eosinophil-targeted therapy: Not the panacea for antibody-mediated rejection? American Journal of Transplantation 2013;13(10):2522-2523. Epub 2013/08/08. doi: 10.1111/ajt.12402. PubMed PMID: 23919601.
  35. 35. Krenzien F, Keshi E, Splith K, Griesel S, Kamali K, Sauer IM, et al. Diagnostic Biomarkers to Diagnose Acute Allograft Rejection After Liver Transplantation: Systematic Review and Meta-Analysis of Diagnostic Accuracy Studies. Front Immunol. 2019;10:758. Epub 2019/04/30. doi: 10.3389/fimmu.2019.00758. PubMed PMID: 31031758; PubMed Central PMCID: PMCPMC6470197.
  36. 36. Aini W, Miyagawa-Hayashino A, Tsuruyama T, Hashimoto S, Sumiyoshi S, Ozeki M, et al. Telomere shortening and karyotypic alterations in hepatocytes in long-term transplanted human liver allografts. Transpl Int. 2012;25(9):956-966. doi: 10.1111/j.1432-2277.2012.01523.x. PubMed PMID: 22775391.

Written By

Tatsuaki Tsuruyama

Submitted: 17 July 2020 Reviewed: 06 October 2020 Published: 10 November 2020