Select your timezone:

Kidney

-

Room: TBD

P8.007 Single Nuclei RNA-sequencing Unravels Complex Renal and Immune Cells Signaling Driving Fibrosis in Kidney Allografts

Jennifer McDaniels, United States

Postdoctoral Fellow
Surgical Sciences
University of Maryland, Baltimore

Abstract

Single nuclei RNA-sequencing unravels complex renal and immune cells signaling driving fibrosis in kidney allografts

Jennifer McDaniels1, Amol Shetty2, Cem Kuscu3, Canan Kuscu3, Elissa Bardhi1, Thomas Rousselle1, James Eason3, Thangamani Muthukumar4, Daniel Maluf5, Valeria Mas1.

1Department of Surgical Sciences, University of Maryland School of Medicine, Baltimore, MD, United States; 2Institute of Genomic Sciences, University of Maryland School of Medicine, Baltimore, MD, United States; 3University of Tennessee Health Science Center, Memphis, TN, United States; 4Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, NY, United States; 5Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States

Introduction: Chronic allograft dysfunction (CAD), characterized by interstitial fibrosis and tubular atrophy (IFTA), is the main cause of late graft loss in kidney transplantation. This study dissects the molecular and cellular heterogeneity of the human kidney stroma and immune cells driving CAD.

Methods: Using single-nuclei RNA sequencing, 41,893 nuclei were evaluated from 8 rare kidney allograft biopsies (6 CAD and 3 normal patients). These needle biopsies were collected on average at ≥15-months posttransplanation and were stored in the cryopreservative, RNAlater. Following, integrative analysis was applied including pathway and enrichment analysis, pseudotime trajectories, and ligand receptor analyses. XY chromosome linked gene expression analysis was also used to determine the origin of both immune and nonimmune cells in sex-mismatch transplants.

Results: Two states of fibrosis were discovered (low and high ECM) that differ based on significant alterations in kidney subclusters, immune cell proportions and phenotypes, and intercellular communication. Pseudotime trajectories revealed a mixed tubule cluster, a key intermediate of injured proximal tubular cells, that transitioned to an activated fibroblast cluster also enriched in myofibroblasts markers. Such differentiation of proximal tubular cells was the main driver of fibrosis. Unique to high ECM, MT1 performed replicative repair evidenced by dedifferentiation and nephrogenic transcriptional signatures. Conversely, low ECM did not show a strong replicative repair signature. Differences in immune cell populations highlighted the dynamic nature of immune responses, some of which, promoted severe metabolic dysfunction and limited repair in low ECM or perpetual tissue regeneration and sustained injury in high ECM. Low ECM was characterized by increased dendritic cells (cDCs, pDCs), mast cells, and monocytes (MO1, MO2) whereas high ECM was characterized by increased B cells, T cells (CD8+ T cells, NKs, and Tregs), and plasma cells. Moreover, 65 ligand-receptor pairs between kidney cells and donor-derived macrophages (previously unidentified after several years post-transplantation) played a novel role in injury propagation.

Conclusion: This work improves our understanding of targetable cell types and regulatory mechanisms involved in tissue repair. Interventions aimed at graft fibrogenesis likely require a more targeted approach based on the unique molecular pathways characterizing each condition.

National Institutes of Health grant R01DK109581. National Institutes of Health grant R01DK122682 (VRM).

Social Media Promotion Image

right-click to download

© 2024 TTS 2022