Tolerogenic dendritic cells: promising cell therapy for acute kidney injury

急性肾损伤 细胞疗法 医学 细胞损伤 树突状细胞 细胞 免疫学 免疫系统 生物 内科学 细胞凋亡 生物化学 遗传学
作者
Y. Nakamura,Tsuyoshi Inoue
出处
期刊:Kidney International [Elsevier]
卷期号:104 (3): 420-422
标识
DOI:10.1016/j.kint.2023.06.015
摘要

There is still no established treatment for acute kidney injury (AKI), and the intervention of AKI remains limited to supportive treatments. Li et al. demonstrated the mechanism by which immune tolerance by dendritic cell ameliorates AKI in a mouse ischemia-reperfusion injury model. The phase I/II clinical trials of tolerogenic dendritic cell therapy have been conducted for kidney transplantation, so it is expected to have potential as a cell therapy for AKI in the future. There is still no established treatment for acute kidney injury (AKI), and the intervention of AKI remains limited to supportive treatments. Li et al. demonstrated the mechanism by which immune tolerance by dendritic cell ameliorates AKI in a mouse ischemia-reperfusion injury model. The phase I/II clinical trials of tolerogenic dendritic cell therapy have been conducted for kidney transplantation, so it is expected to have potential as a cell therapy for AKI in the future. Dendritic cells (DCs), discovered by Dr. Ralph M. Steinman and Dr. Zanvil A. Cohn in 1973, who awarded the Nobel Prize in Physiology or Medicine in 2011, are antigen-presenting cells and play a pivotal role in initiating immune response of acquired immunity. Precursor cells of DCs produced in the bone marrow are widely distributed in peripheral tissues as immature DCs. Immature DCs phagocytose antigens and, on activation, migrate to their lymph nodes, where they form major histocompatibility complex (MHC) molecules with antigen-derived peptides, which activate T cells and induce antigen-specific immune responses. DCs are not only more potent than other antigen-presenting cells but also have the ability to activate naïve T cells, induce differentiation of T cells by different costimulatory factors and induce immune tolerance in response to environmental factors. DCs with such diverse functions are formed from a heterogeneous cell population, consisting of multiple subsets with distinct phenotypes and functions. DC subsets can be broadly categorized into conventional DCs, which primarily promote immune responses and facilitate the differentiation and induction of antigen-specific T cells, and plasmacytoid DCs, which produce interferons during viral infections and contribute to the host defense in antiviral immunity. On the other hand, DCs with an immature phenotype, known as tolerogenic DCs (tolDCs), do not induce antigen-specific immune responses and instead exert immunosuppressive functions.1Lin J. Wang H. Liu C. et al.Dendritic cells: versatile players in renal transplantation.Front Immunol. 2021; 12654540Google Scholar DCs induce immune responses when they express high levels of MHC and costimulatory molecules CD80/CD86 and produce interleukin-12 (IL-12). On the other hand, when DCs exhibit low expression of MHC and costimulatory molecules and produce immunosuppressive cytokines such as IL-10, they induce immune tolerance.2Zahorchak A.F. Macedo C. Hamm D.E. et al.High PD-L1/CD86 MFI ratio and IL-10 secretion characterize human regulatory dendritic cells generated for clinical testing in organ transplantation.Cell Immunol. 2018; 323: 9-18Crossref PubMed Scopus (32) Google Scholar Therefore, tolDCs are regarded as promising therapeutic approaches for autoimmune diseases and allograft rejection in transplantation. In these diseases, the use of immunosuppressive drugs poses issues such as increased susceptibility to infections, side effects, and the risk of cancer. However, cellular therapy with tolDCs allows for a reduction in the amounts of immunosuppressive drugs used. Phase I/II clinical trials using tolDCs have been conducted for autoimmune diseases such as type 1 diabetes, rheumatoid arthritis, Crohn disease, multiple sclerosis, and liver and kidney transplantation.3Thomson A.W. Metes D.M. Ezzelarab M.B. et al.Regulatory dendritic cells for human organ transplantation.Transplant Rev (Orlando). 2019; 33: 130-136Crossref PubMed Scopus (38) Google Scholar,4Sawitzki B. Harden P.N. Reinke P. et al.Regulatory cell therapy in kidney transplantation (The ONE Study): a harmonised design and analysis of seven non-randomised, single-arm, phase 1/2A trials.Lancet. 2020; 395: 1627-1639Abstract Full Text Full Text PDF PubMed Scopus (210) Google Scholar The generation of tolDCs used in ex vivo cell therapy involves inducing tolerance in bone marrow–derived progenitors (in animals) or CD14+ monocytes from peripheral blood mononuclear cells (in humans) using drugs such as vitamin D, IL-10, and dexamethasone. Many clinical trials have demonstrated high tolerability, with no observed treatment-related adverse reactions. Regarding kidney transplantation, it has been reported that infusion of donor-derived tolDC before transplantation interacts with cytotoxic T lymphocyte–associated antigen 4 present on host T cells, extending the survival period of the transplant.5Ezzelarab M.B. Lu L. Shufesky W.F. et al.Donor-derived regulatory dendritic cell infusion maintains donor-reactive CD4(+)CTLA4(hi) T cells in non-human primate renal allograft recipients treated with CD28 co-stimulation blockade.Front Immunol. 2018; 9: 250Crossref PubMed Scopus (21) Google Scholar However, the detailed mechanism is not yet fully understood. In the paper by Li et al.6Li J.S.Y. Robertson H. Trinh K. et al.Tolerogenic dendritic cells protect against acute kidney injury.Kidney Int. 2023; 104: 492-507Google Scholar in this issue, the immunoregulatory mechanisms of tolDCs were examined, particularly their potential to improve acute kidney injury (AKI), and their impact on kidney tubular epithelial cells (RTECs) was investigated (Figure 1). The authors evaluated the impact of ex vivo DC cell therapy on AKI using a bilateral kidney ischemia-reperfusion injury (IRI) mouse model. Initially, bone marrow–derived DCs and tolDCs (±lipopolysaccharide [LPS]) were induced through exposure to IL-10/1α,25-dihydroxyvitamin D3, which was previously used in clinical trials of liver and kidney transplantation. Analysis of expression markers confirmed the induction of tolDCs based on the downregulation of MHC II, CD80, CD86, an increase in PD-L1:CD86 mean fluorescence intensity ratio, and secretion of IL-10. Subsequently, in vitro coculture experiments were conducted with induced tolDCs and LPS-tolDCs in a noncontact manner with RTECs, followed by LPS-induced injury. The presence of LPS-tolDCs attenuated the impairment of tumor necrosis factor-α, lipocalin-2, and kidney injury molecule-1 in RTECs. The induced tolDCs and LPS-tolDCs exhibited not only inhibitory effects on lymphocyte proliferation but also an increase in PD-L1:CD86 mean fluorescence intensity ratio, enhanced IL-10 production, and suppression of IL-12p70 (proinflammatory factor) production. Furthermore, in vivo transplantation of tolDCs was performed in IRI mice. Administration of LPS-tolDCs before injury mitigated the elevation of serum creatinine levels, histological damage, and cell death caused by IRI. The kidney protective effect was also observed on the administration of allogenic-tolDCs, which were generated by conditioning DCs from mice of different strains. However, tolDCs without LPS stimulation did not show a significant protective effect against kidney injury. Both LPS-tolDCs and allogenic-tolDCs showed common changes in markers associated with tolerance induction. This result suggests the potential for organ protection even in the context of allogeneic transplantation. The authors suggested that the differential gene expression of chemokines such as CC-chemokine receptor 7 and CC-chemokine receptor 4, which were higher in LPS-tolDCs compared with unstimulated tolDCs, may contribute to the kidney-protective effect by facilitating the retention of LPS-tolDCs in the kidney. LPS-tolDCs exhibited higher immune activation and cytokine production pathways compared with tolDCs, suggesting that LPS-tolDCs possess the characteristics of alternatively activated DCs. Alternatively activated DCs are tolDCs that are selectively activated by inflammatory stimuli such as exposure to LPS. Although the effects of IL-10–conditioned tolDCs are modest, alternatively activated DCs exert significant effects in controlling inflammatory immune responses in vivo, particularly in protecting against graft-versus-host disease in transplantation.7Morelli A.E. Thomson A.W. Tolerogenic dendritic cells and the quest for transplant tolerance.Nat Rev Immunol. 2007; 7: 610-621Crossref PubMed Scopus (748) Google Scholar Furthermore, the spatial transcriptomics analysis revealed not only the suppression of proinflammatory cytokine expression after LPS-tolDC therapy but also the potential to alleviate tubular injury by promoting proximal tubular lipid oxidation and fatty acid metabolism. This result could explain the protective effect of tolDC on RTECs. Even more, these responses persisted even after depletion of recipient macrophages. What is interesting about this work by Li et al. is that they not only evaluated the impact of tolDC cell therapy on AKI for the first time but also demonstrated that the adoptive cell therapy of tolDCs had no influence on T cells or other myeloid cell populations, but rather it proved to suppress the damage to RTECs. There are reports suggesting that in renal IRI, proinflammatory cytokines and tumor necrosis factor derived from hypoxic endothelial cells can recruit DCs, and subsequently, hypoxia-inducible factor 1a induces the maturation of DCs, leading to impaired kidney function.8Jantsch J. Chakravortty D. Turza N. et al.Hypoxia and hypoxia-inducible factor-1 alpha modulate lipopolysaccharide-induced dendritic cell activation and function.J Immunol. 2008; 180: 4697-4705Crossref PubMed Scopus (318) Google Scholar,9Xu L. Sharkey D. Cantley L.G. Tubular GM-CSF promotes late MCP-1/CCR2-mediated fibrosis and inflammation after ischemia/reperfusion injury.J Am Soc Nephrol. 2019; 30: 1825-1840Crossref PubMed Scopus (72) Google Scholar Further investigation is needed regarding the effects of ex vivo tolDC cell therapy on renal resident DCs and other immune cells. On the other hand, spatial transcriptomic analysis currently has limited resolution, making it challenging to fully elucidate the precise spatial relationships and cell-cell interactions within the complex and diverse organ such as the kidney. In particular, the interaction between macrophages and proximal tubules, which is the focus of this study, requires further investigation beyond the findings of this experiment alone. Furthermore, in order to bring tolDC ex vivo cell therapy into actual clinical practice, further experiments in other AKI models are awaited. In summary, the work by Li et al. demonstrates that tolDC cell therapy can ameliorate kidney injury in a mouse model of IRI. The beneficial effects of tolDC therapy were also observed in in vitro experiments where tolDCs showed a reduction in kidney injury markers in RTECs without direct contact. Spatial transcriptomic analysis suggested that tolDCs might alleviate kidney injury by normalizing lipid oxidation and fatty acid metabolism pathways in the proximal tubules. The accumulation of further knowledge regarding the effectiveness and mechanisms of tolDCs in AKI has the potential for tolDC cell therapy to become a promising treatment for AKI. All the authors declared no competing interests. Tolerogenic dendritic cells protect against acute kidney injuryKidney InternationalVol. 104Issue 3PreviewIschemia reperfusion injury is a common precipitant of acute kidney injury that occurs following disrupted perfusion to the kidney. This includes blood loss and hemodynamic shock, as well as during retrieval for deceased donor kidney transplantation. Acute kidney injury is associated with adverse long-term clinical outcomes and requires effective interventions that can modify the disease process. Immunomodulatory cell therapies such as tolerogenic dendritic cells remain a promising tool, and here we tested the hypothesis that adoptively transferred tolerogenic dendritic cells can limit kidney injury. Full-Text PDF Open Access

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
mm完成签到,获得积分20
1秒前
zhangzhang完成签到,获得积分10
1秒前
linfordlu完成签到,获得积分0
1秒前
安安完成签到,获得积分20
1秒前
今后应助孙佳琦采纳,获得10
1秒前
CodeCraft应助顾化蛹采纳,获得10
2秒前
3秒前
甜甜清发布了新的文献求助10
4秒前
Akim应助嘎嘎嘎采纳,获得10
4秒前
完美世界应助小乔同学采纳,获得10
4秒前
瑶瑶要加油完成签到,获得积分10
4秒前
5秒前
哭泣慕晴完成签到 ,获得积分10
5秒前
Tapioca完成签到,获得积分10
5秒前
feng完成签到,获得积分10
6秒前
隐形曼青应助顶刊我来了采纳,获得10
6秒前
stars发布了新的文献求助10
6秒前
6秒前
8秒前
Kiki发布了新的文献求助10
8秒前
123456完成签到,获得积分10
9秒前
记忆关注了科研通微信公众号
9秒前
huohua完成签到,获得积分10
9秒前
10秒前
10秒前
黄龙光完成签到 ,获得积分10
10秒前
细菌学家完成签到 ,获得积分10
10秒前
完美世界应助yey采纳,获得10
10秒前
77z完成签到,获得积分10
11秒前
superming发布了新的文献求助10
12秒前
13秒前
zhangzhang发布了新的文献求助10
13秒前
abc123发布了新的文献求助30
13秒前
黄龙光关注了科研通微信公众号
14秒前
彭于晏应助bb采纳,获得10
14秒前
新明发布了新的文献求助10
14秒前
领导范儿应助虚幻采枫采纳,获得10
15秒前
嘎嘎嘎发布了新的文献求助10
15秒前
15秒前
杨心茹应助半生瓜711321采纳,获得10
16秒前
高分求助中
Handbook of Fuel Cells, 6 Volume Set 1666
Floxuridine; Third Edition 1000
Tracking and Data Fusion: A Handbook of Algorithms 1000
Sustainable Land Management: Strategies to Cope with the Marginalisation of Agriculture 800
消化器内視鏡関連の偶発症に関する第7回全国調査報告2019〜2021年までの3年間 500
One Man Talking: Selected Essays of Shao Xunmei, 1929–1939 500
Framing China: Media Images and Political Debates in Britain, the USA and Switzerland, 1900-1950 500
热门求助领域 (近24小时)
化学 医学 生物 材料科学 工程类 有机化学 生物化学 内科学 物理 纳米技术 计算机科学 化学工程 复合材料 基因 遗传学 催化作用 物理化学 免疫学 冶金 细胞生物学
热门帖子
关注 科研通微信公众号,转发送积分 2861490
求助须知:如何正确求助?哪些是违规求助? 2466871
关于积分的说明 6688461
捐赠科研通 2158099
什么是DOI,文献DOI怎么找? 1146415
版权声明 585109
科研通“疑难数据库(出版商)”最低求助积分说明 563292