リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「B cell depletion with anti-CD20 mAb exacerbates anti-donor CD4+ T cell responses in highly sensitized transplant recipients」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

B cell depletion with anti-CD20 mAb exacerbates anti-donor CD4+ T cell responses in highly sensitized transplant recipients

田中 飛鳥 広島大学

2021.11.25

概要

www.nature.com/scientificreports

OPEN

B cell depletion with anti‑CD20
mAb exacerbates anti‑donor ­CD4+
T cell responses in highly sensitized
transplant recipients
Asuka Tanaka, Kentaro Ide*, Yuka Tanaka, Masahiro Ohira, Hiroyuki Tahara &
Hideki Ohdan*
Pretransplant desensitization with rituximab has been applied to preformed donor-specific antihuman leukocyte antigen antibody (DSA)-positive recipients for elimination of preformed DSA. We
investigated the impact of pretransplant desensitization with rituximab on anti-donor T cell responses
in DSA-positive transplant recipients. To monitor the patients’ immune status, mixed lymphocyte
reaction (MLR) assays were performed before and after desensitization with rituximab. Two weeks
after rituximab administration, the stimulation index (SI) of anti-donor ­CD4+ T cells was significantly
higher in the DSA-positive recipients than in the DSA-negative recipients. To investigate the
mechanisms of anti-donor hyper responses of ­CD4+ T cells after B cell depletion, highly sensitized mice
models were injected with anti-CD20 mAb to eliminate B cells. Consistent with clinical observations,
the SI values of anti-donor ­CD4+ T cells were significantly increased after anti-CD20 mAb injection
in the sensitized mice models. Adding B cells isolated from untreated sensitized mice to MLR
significantly inhibited the enhancement of anti-donor ­CD4+ T cell response. The depletion of the ­CD5+
B cell subset, which exclusively included IL-10-positive cells, from the additive B cells abrogated such
inhibitory effects. These findings demonstrate that IL-10+ ­CD5+ B cells suppress the excessive response
of anti-donor ­CD4+ T cells responses in sensitized recipients.
It has been reported that preformed DSAs are associated with detrimental effects in transplant r­ ecipients1. In
kidney transplantation, preformed DSAs, regardless of whether they are HLA-class I or II, can trigger hyper
acute, accelerated acute, and early acute antibody-mediated rejection (ABMR)2, 3. Acute and chronic ABMR are
major factors of renal allograft dysfunction and l­oss4. In contrast to other types of organ transplantation, liver
transplant (LT) recipients are considered resistant to ABMR caused by D
­ SAs5, 6. However, recent studies suggest
that liver allografts have a relative resistance to ABMR, but specific situations can override the liver’s natural
resistance and defense m
­ echanisms7, 8. In detail, preformed class I and/or II DSAs, detected by single-antigen
bead analyses, with a mean fluorescence intensity (MFI) ≥ 5000, are independently correlated with a greater risk
of ­death9. Furthermore, preformed class I DSA (MFI ≥ 5000) also disproportionately affects patients transplanted
with a high calculated model for end-stage liver disease (MELD) score and those who received lower quality
organs [donor risk index (DRI) > 1.5]10.
To eliminate the preformed DSAs, several desensitization protocols comprising plasmapheresis, splenectomy,
intravenous immunoglobulins (IVIG), and/or anti-B cell immunosuppressant treatment in the recipients have
been reported for successful ­transplant11–13. Among these, B cell depletion with the prophylactic use of rituximab
has also been applied in preformed DSA-positive r­ ecipients14, 15. However, depletion of B cells may influence T
cell allo-responses because B cells are effective antigen-presenting cells that can activate allo-specific T c­ ells16.
The cytokine release syndrome induced by rituximab may also enhance T cell ­activation17. Reportedly, rituximab
can modulate the immunoresponse by secretion of IL-10 and macrophage inflammatory protein-1β18. However,
few studies have clearly investigated whether immune modification after administration of rituximab promotes
or inhibits T cell allo-response, even though T cells are also sensitized with alloantigens in DSA-positive sensitized patients.

Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences,
Hiroshima University, 1‑2‑3 Kasumi Minami‑ku, Hiroshima 734‑8551, Japan. *email: ideken@hiroshima-u.ac.jp;
hohdan@hiroshima-u.ac.jp
Scientific Reports |

(2021) 11:18180

| https://doi.org/10.1038/s41598-021-97748-9

1
Vol.:(0123456789)

www.nature.com/scientificreports/

Recipient age, years, median (range)

ABO-incompatible (n = 45)

DSA-positive (n = 17)

53.0 (20–71)

61.0 (24–70)

Gender

P value
0.50
< 0.01

Male

31

3

Female

14

14

Kidney

32

11

Liver

13

6

Donor age, years, median (range)

55.0 (23–78)

58.0 (28–74)

Organ

0.63

Gender

0.43
< 0.01

Male

17

14

Female

28

3

Child

9

4

Parent

8

3

Sibling

2

1

Spouse

26

9

Relationship

0.98

HLA allele MM
A (0: 1: 2)

10: 19: 16

3: 9: 5

0.75

B (0: 1: 2)

3: 22: 20

0: 8: 9

0.51

DR (0: 1: 2)

7: 21: 17

0: 12: 5

0.12

Table 1.  Characteristics of ABO-incompatible and DSA-positive patients. A difference was considered
significant if the p-value was <0.05.

We desensitized DSA-positive patients with rituximab and plasmapheresis, which was adopted from a protocol for ABO-blood type incompatible (ABO-I) transplant r­ ecipients19. We have recently shown that pretransplant
desensitization with rituximab has a minimal effect on the alloreactive T cell responses by comparing ABO-I
and ABO-compatible g­ roups20. However, the previous study excluded DSA-positive recipients who might have
preformed donor-reactive T cells. Hence, the objective of this study was to elucidate the impact of pretransplant
desensitization with rituximab on the subsequent response of T cells to donor-antigens in DSA-positive transplant recipients. Moreover, a highly sensitized murine model was applied to investigate the mechanisms of the
significant impacts of B cell depletion by injecting anti-CD20 monoclonal antibodies (mAbs) on anti-donor T
cell responses.

Results

T cell immune responses before and after rituximab administration in desensitized
patients.  The baseline characteristics of the desensitized patients are listed in Table 1.

Of these, 17 patients were DSA-positive (kidney, n = 11; liver, n = 6) and the remaining 45 were DSA-negative
ABO-I patients (kidney, n = 32; liver, n = 13). Of the DSA-positive group, 9 patients were CDC cross match
(XM)-positive (kidney, n = 5; liver, n = 4), and the remaining eight were CDC XM-negative flow cytometry
crossmatch (FCXM)-positive patients (kidney, n = 6; liver, n = 2). In the DSA-positive group, there were more
female recipients than males, and male donors than females. Similar to our previous ­report20, the proportion of
peripheral blood I­ gM+ ­CD19+ B cells in all the recipients decreased below 0.1% at 2 weeks after administration of
rituximab (data not shown). To evaluate the T cell immune status of recipients, we performed mixed lymphocyte
reaction (MLR) assays using a carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeling technique before
rituximab administration and 2 weeks after rituximab administration (before transplantation). There were no
significant differences in the SI values for the C
­ D4+ T cell responses between the two groups before rituximab
administration (Fig. 1A,C).
Of note, after rituximab administration, the SI values for the ­CD4+ T cell responses to donor stimulation
were significantly higher in the DSA-positive group than those in the ABO-I group (p < 0.05, Fig. 1A,C). There
were no significant differences in the SI values for the ­CD8+ T cell responses to donor between the two groups
(Fig. 1B,D). After transplantation, kidney transplant recipients did not suffer from T cell-mediated rejection
(TCMR), probably because they received either rabbit anti-thymocyte globulin (ATG) or basiliximab as induction therapy. In contrast, two out of five DSA-positive liver transplant recipients suffered from severe TCMR
requiring rabbit ATG treatment.

Kinetics of anti‑allo Ab titers of a highly sensitized murine model.  To investigate the mechanisms

of anti-donor hyper responses of ­CD4+ T cells after B cell depletion, naïve Balb/c mice were transplanted with
C57BL/6 skin grafts to prepare a highly sensitized model, and the sensitized mice were injected with anti-CD20
mAb to eliminate B cells. To confirm allo-sensitization by allogenic skin transplantation, the production of anti-

Scientific Reports |
Vol:.(1234567890)

(2021) 11:18180 |

https://doi.org/10.1038/s41598-021-97748-9

2

www.nature.com/scientificreports/

Figure 1.  T cell immune responses before and after rituximab administration in desensitized patients. Sixtytwo patients received pretransplant desensitization with rituximab. Of these, 17 patients were DSA-positive
(kidney, n = 11; liver, n = 6) and the remaining 45 were DSA-negative ABO-I patients (kidney, n = 32; liver,
n = 13). To evaluate the T cell immune status of recipients, CFSE-MLR assays were performed before rituximab
administration and 2 weeks after rituximab administration (before transplantation). The stimulation index (SI)
values of each ­CD4+ T cells (A, C) and ­CD8+ T cells (B, D) before and after rituximab are shown. White box,
ABO-I group; gray box, DSA-positive group. Data are shown as median, 25th and 75th percentiles, and range.
The Wilcoxon-Mann–Whitney test was used to evaluate differences between before and after desensitization.
C57BL/6 Abs in the sera of recipient Balb/c mice was serially evaluated. The levels of anti-C57BL/6 IgG titers
gradually increased and reached a peak at nine weeks after the first skin transplantation (Figs. 2 and S1).
In the anti-CD20 mAb group (n = 5), unlike clinical desensitization therapy, plasma exchange was not performed in mouse experiments, and thus no dramatic decrease in anti-C57BL/6 IgG titers was observed. Instead,
titers gradually decreased after injection of anti-CD20 mAb at week 6 after the first skin transplantation. The
differences in anti-C57BL/6 IgG titers between the groups reached statistical significance at week 12 (p < 0.05). ...

この論文で使われている画像

参考文献

1. Patel, R. & Terasaki, P. I. Significance of the positive crossmatch test in kidney transplantation. N. Engl. J. Med. 280, 735–739.

https://​doi.​org/​10.​1056/​NEJM1​96904​03280​1401 (1969).

2. Lefaucheur, C. et al. Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J. Am. Soc. Nephrol.

21, 1398–1406. https://​doi.​org/​10.​1681/​ASN.​20091​01065 (2010).

3. Honger, G. et al. Pretransplant IgG subclasses of donor-specific human leukocyte antigen antibodies and development of antibodymediated rejection. Transplantation 92, 41–47. https://​doi.​org/​10.​1097/​TP.​0b013​e3182​1cdf0d (2011).

4. Cornell, L. D. Renal allograft pathology in the sensitized patient. Curr. Opin. Organ Transplant. 18, 327–336. https://​doi.​org/​10.​

1097/​MOT.​0b013​e3283​614c5a (2013).

5. Iwatsuki, S., Rabin, B. S., Shaw, B. W. Jr. & Starzl, T. E. Liver transplantation against T cell-positive warm crossmatches. Transplant.

Proc. 16, 1427–1429 (1984).

6. Gordon, R. D. et al. The antibody crossmatch in liver transplantation. Surgery 100, 705–715 (1986).

7. Goh, A., Scalamogna, M., De Feo, T., Poli, F. & Terasaki, P. I. Human leukocyte antigen crossmatch testing is important for liver

retransplantation. Liver Transplant. 16, 308–313. https://​doi.​org/​10.​1002/​lt.​21981 (2010).

Scientific Reports |

(2021) 11:18180 |

https://doi.org/10.1038/s41598-021-97748-9

Vol.:(0123456789)

www.nature.com/scientificreports/

8. Kaneku, H. et al. Donor-specific human leukocyte antigen antibodies of the immunoglobulin G3 subclass are associated with

chronic rejection and graft loss after liver transplantation. Liver Transplant. 18, 984–992. https://​doi.​org/​10.​1002/​lt.​23451 (2012).

9. O’Leary, J. G. et al. Preformed class II donor-specific antibodies are associated with an increased risk of early rejection after liver

transplantation. Liver Transplant. 19, 973–980. https://​doi.​org/​10.​1002/​lt.​23687 (2013).

10. Demetris, A. J., Zeevi, A. & O’Leary, J. G. ABO-compatible liver allograft antibody-mediated rejection: an update. Curr. Opin.

Organ Transplant. 20, 314–324. https://​doi.​org/​10.​1097/​MOT.​00000​00000​000194 (2015).

11. Glotz, D. et al. Desensitization and subsequent kidney transplantation of patients using intravenous immunoglobulins (IVIg). Am.

J. Transplant. 2, 758–760 (2002).

12. Jordan, S. C. et al. Evaluation of intravenous immunoglobulin as an agent to lower allosensitization and improve transplantation

in highly sensitized adult patients with end-stage renal disease: report of the NIH IG02 trial. J. Am. Soc. Nephrol. 15, 3256–3262.

https://​doi.​org/​10.​1097/​01.​ASN.​00001​45878.​92906.​9F (2004).

13. Stegall, M. D., Gloor, J., Winters, J. L., Moore, S. B. & Degoey, S. A comparison of plasmapheresis versus high-dose IVIG desensitization in renal allograft recipients with high levels of donor specific alloantibody. Am. J. Transplant. 6, 346–351. https://​doi.​org/​

10.​1111/j.​1600-​6143.​2005.​01178.x (2006).

14. Vieira, C. A. et al. Rituximab for reduction of anti-HLA antibodies in patients awaiting renal transplantation: 1. Safety, pharmacodynamics, and pharmacokinetics. Transplantation 77, 542–548 (2004).

15. Vo, A. A. et al. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N. Engl. J. Med. 359,

242–251. https://​doi.​org/​10.​1056/​NEJMo​a0707​894 (2008).

16. Janeway, C. A. Jr., Ron, J. & Katz, M. E. The B cell is the initiating antigen-presenting cell in peripheral lymph nodes. J. Immunol.

138, 1051–1055 (1987).

17. Clatworthy, M. R. et al. B-cell-depleting induction therapy and acute cellular rejection. N. Engl. J. Med. 360, 2683–2685. https://​

doi.​org/​10.​1056/​NEJMc​08084​81 (2009).

18. Kamburova, E. G. et al. Cytokine release after treatment with rituximab in renal transplant recipients. Transplantation 99, 1907–

1911. https://​doi.​org/​10.​1097/​TP.​00000​00000​000515 (2015).

19. Tanimine, N. et al. Kinetics of cellular and humoral immunity in a successful case of positive crossmatch kidney transplantation:

a case report. Transplant. Proc. 43, 2411–2414. https://​doi.​org/​10.​1016/j.​trans​proce​ed.​2011.​05.​039 (2011).

20. Morimoto, H. et al. Different sensitivity of rituximab-treatment to B-cells between ABO-incompatible kidney and liver transplantation. Hum. Immunol. 77, 456–463. https://​doi.​org/​10.​1016/j.​humimm.​2016.​04.​013 (2016).

21. Hirose, T. et al. PD-L1/PD-L2-expressing B-1 cells inhibit alloreactive T cells in mice. PLoS ONE 12, e0178765. https://​doi.​org/​

10.​1371/​journ​al.​pone.​01787​65 (2017).

22. Marino, J. et al. B cell depletion with an anti-CD20 antibody enhances alloreactive memory T cell responses after transplantation.

Am. J. Transplant. 16, 672–678. https://​doi.​org/​10.​1111/​ajt.​13483 (2016).

23. Marino, J., Gonzalez-Nolasco, B., Wang, X., Orent, W. & Benichou, G. Contrasting effects of B cell depletion on CD4(+) and

CD8(+) memory T cell responses generated after transplantation. Am. J. Transplant. https://​doi.​org/​10.​1111/​ajt.​15858 (2020).

24. Lanzavecchia, A. Antigen-specific interaction between T and B cells. Nature 314, 537–539 (1985).

25. Tahara, H., Ide, K., Basnet, N., Tanaka, Y. & Ohdan, H. Determination of the precursor frequency and the reaction intensity of

xenoreactive human T lymphocytes. Xenotransplantation 17, 188–196. https://​doi.​org/​10.​1111/j.​1399-​3089.​2010.​00575.x (2010).

26. Yanaba, K. et al. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory

responses. Immunity 28, 639–650. https://​doi.​org/​10.​1016/j.​immuni.​2008.​03.​017 (2008).

27. Rosser, E. C. & Mauri, C. Regulatory B cells: origin, phenotype, and function. Immunity 42, 607–612. https://​doi.​org/​10.​1016/j.​

immuni.​2015.​04.​005 (2015).

28. Lal, G. et al. Interleukin-10 from marginal zone precursor B-cell subset is required for costimulatory blockade-induced transplantation tolerance. Transplantation 99, 1817–1828. https://​doi.​org/​10.​1097/​TP.​00000​00000​000718 (2015).

29. Dasgupta, S., Dasgupta, S. & Bandyopadhyay, M. Regulatory B cells in infection, inflammation, and autoimmunity. Cell Immunol.

352, 104076. https://​doi.​org/​10.​1016/j.​celli​mm.​2020.​104076 (2020).

30. Madan, R. et al. Nonredundant roles for B cell-derived IL-10 in immune counter-regulation. J. Immunol. 183, 2312–2320. https://​

doi.​org/​10.​4049/​jimmu​nol.​09001​85 (2009).

31. Ide, K. et al. A phased desensitization protocol with rituximab and bortezomib for highly sensitized kidney transplant candidates.

Transplant. Direct 1, e17. https://​doi.​org/​10.​1097/​TXD.​00000​00000​000526 (2015).

32. Tanaka, Y., Ohdan, H., Onoe, T. & Asahara, T. Multiparameter flow cytometric approach for simultaneous evaluation of proliferation and cytokine-secreting activity in T cells responding to allo-stimulation. Immunol. Investig. 33, 309–324 (2004).

33. Ohdan, H. Quantification of T-cell proliferation for individualizing immunosuppressive therapy for transplantation patients. Clin.

Pharmacol. Ther. 87, 23–26. https://​doi.​org/​10.​1038/​clpt.​2009.​171 (2010).

Acknowledgements

We thank Onoe T, Morimoto H, Shimizu S, and Tanimine N, Taguchi K for advice and encouragement, and

Kashihara M and Piao J for their technical assistance. This work was carried out in part at the Research Facilities for Laboratory Animal Science, Natural Science Center for Basic Research and Development (N-BARD),

Hiroshima University. This work was supported by JSPS KAKENHI (grant numbers JP17K10512, JP20K08982)

and AMED (grant number JP20ek0510029).

Author contributions

Participated in research design: A.T., K.I., H.O. Participated in the writing of the paper: A.T., K.I., H.O. Participated in the performance of the research: A.T., K.I., Y.T., M.O., H.T. Participated in data analysis: A.T., K.I.,

Y.T., H.O.

Competing interests The authors declare no competing interests.

Additional information

Supplementary Information The online version contains supplementary material available at https://​doi.​org/​

10.​1038/​s41598-​021-​97748-9.

Correspondence and requests for materials should be addressed to K.I. or H.O.

Reprints and permissions information is available at www.nature.com/reprints.

Scientific Reports |

Vol:.(1234567890)

(2021) 11:18180 |

https://doi.org/10.1038/s41598-021-97748-9

10

www.nature.com/scientificreports/

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and

institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International

License, which permits use, sharing, adaptation, distribution and reproduction in any medium or

format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the

Creative Commons licence, and indicate if changes were made. The images or other third party material in this

article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the

material. If material is not included in the article’s Creative Commons licence and your intended use is not

permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.

© The Author(s) 2021

Scientific Reports |

(2021) 11:18180 |

https://doi.org/10.1038/s41598-021-97748-9

11

Vol.:(0123456789)

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る