ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Please note that you are viewing an archived section from 2022 and some content may be unavailable. To unlock all content for 2022, please visit the archives.

Abstract: FR-PO173

Role of Double-Negative T Cells in Repair After Experimental Severe AKI

Session Information

  • AKI: Mechanisms - II
    November 04, 2022 | Location: Exhibit Hall, Orange County Convention Center‚ West Building
    Abstract Time: 10:00 AM - 12:00 PM

Category: Acute Kidney Injury

  • 103 AKI: Mechanisms

Authors

  • Lee, Kyungho, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
  • Gharaie, Sepideh, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
  • Kurzhagen, Johanna T., Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
  • Arend, Lois J., Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
  • Noel, Sanjeev, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
  • Rabb, Hamid, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
Background

T cells mediate organ injury and repair. A proportion of kidney αβ T cells are unconventional double-negative (DN) T cells (CD4- CD8-), which have anti-inflammatory properties previously demonstrated to protect from early injury in AKI. However, their role in repair from AKI has not been studied. We aimed to elucidate the role of DN T cells in AKI repair.

Methods

C57B6 mice underwent unilateral ischemia-reperfusion injury (IRI) surgery with 40 min ischemia. DN T cells, isolated from Faslgld mouse lymph nodes, were adoptively transferred AFTER IRI. Vehicle (neg. control) or Tregs (pos. control, Kidney Int 2009 PMID: 19625990) were also injected. GFR was measured by FITC-sinistrin-based method. Fibrosis was assessed with Masson trichrome staining. Profibrotic genes were measured with quantitative RT-PCR. T cells from postischemic kidneys were studied by flow cytometry.

Results

Percentages and the numbers of DN T cells markedly decreased in postischemic kidneys at 3 weeks from IRI compared to normal kidneys (18.7±1.1 vs 6.9±0.5% of αβ T cells, P<.001, 5.9±0.6×104 vs 0.9±0.3×104 cells/kidney, P<.001). DN T cell CD44 (95.0±0.4 vs 90.8±0.7%, P<.001) and CD69 (94.1±0.3 vs 85.8±1.1%, P<.001) were reduced, while DN T immune checkpoint TIGIT (0.6±0.2 vs 2.5±0.5%, P=.005) and NK1.1 (35.9±2.6 vs 52.4±3.5%, P=.004) were upregulated. Post-AKI transfer of DN T cells improved renal recovery, as did Treg transfer, with 3 week-GFR of 1176±27 μL/min/100g (vehicle, 1076±25, P=.001; Treg 1217±42, P=.399) and outer medullary fibrosis of 64.5±2.3% (vehicle, 82.0±1.9, P<.001; Treg 45.4±9.3%, P=.058). Tgfb1 was lower in the DN T cell transfer group (1.0±0.1 fold) than vehicle group (vehicle, 1.3±0.1, P=.012; Treg, 0.8±0.1, P=.226). Postischemic kidneys from DN T cell transferred mice had less effector memory CD4 T cells (94.7±0.2%) compared to those from vehicle group (vehicle, 96.9±0.3, P<.001; Treg 93.7±0.3%, P=.021). DN T cell transfer enhanced tubular proliferation, with higher Ki67 expression of CD45- E-cadherin+ cells (73.4±1.1; vehicle, 68.5±1.2, P=.011; Treg, 71.8±2.2%, P=.304).

Conclusion

Kidney DN T cells undergo quantitative and phenotypical changes long-term after severe ischemic kidney injury. Post-injury infusions of DN T cells, like T regs, can hasten repair and decrease fibrosis. A potential mechanism is by regulating kidney CD4 T cells.

Funding

  • NIDDK Support