Determining clinically relevant features in cytometry data using persistent homology.

Cytometry experiments yield high-dimensional point cloud data that is difficult to interpret manually. Boolean gating techniques coupled with comparisons of relative abundances of cellular subsets is the current standard for cytometry data analysis. However, this approach is unable to capture more s...

Full description

Bibliographic Details
Main Authors: Soham Mukherjee, Darren Wethington, Tamal K Dey, Jayajit Das
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2022-03-01
Series:PLoS Computational Biology
Online Access:https://doi.org/10.1371/journal.pcbi.1009931
_version_ 1797984427490410496
author Soham Mukherjee
Darren Wethington
Tamal K Dey
Jayajit Das
author_facet Soham Mukherjee
Darren Wethington
Tamal K Dey
Jayajit Das
author_sort Soham Mukherjee
collection DOAJ
description Cytometry experiments yield high-dimensional point cloud data that is difficult to interpret manually. Boolean gating techniques coupled with comparisons of relative abundances of cellular subsets is the current standard for cytometry data analysis. However, this approach is unable to capture more subtle topological features hidden in data, especially if those features are further masked by data transforms or significant batch effects or donor-to-donor variations in clinical data. We present that persistent homology, a mathematical structure that summarizes the topological features, can distinguish different sources of data, such as from groups of healthy donors or patients, effectively. Analysis of publicly available cytometry data describing non-naïve CD8+ T cells in COVID-19 patients and healthy controls shows that systematic structural differences exist between single cell protein expressions in COVID-19 patients and healthy controls. We identify proteins of interest by a decision-tree based classifier, sample points randomly and compute persistence diagrams from these sampled points. The resulting persistence diagrams identify regions in cytometry datasets of varying density and identify protruded structures such as 'elbows'. We compute Wasserstein distances between these persistence diagrams for random pairs of healthy controls and COVID-19 patients and find that systematic structural differences exist between COVID-19 patients and healthy controls in the expression data for T-bet, Eomes, and Ki-67. Further analysis shows that expression of T-bet and Eomes are significantly downregulated in COVID-19 patient non-naïve CD8+ T cells compared to healthy controls. This counter-intuitive finding may indicate that canonical effector CD8+ T cells are less prevalent in COVID-19 patients than healthy controls. This method is applicable to any cytometry dataset for discovering novel insights through topological data analysis which may be difficult to ascertain otherwise with a standard gating strategy or existing bioinformatic tools.
first_indexed 2024-04-11T07:02:40Z
format Article
id doaj.art-a8882e979b7d4715acc9a9a6eec8a863
institution Directory Open Access Journal
issn 1553-734X
1553-7358
language English
last_indexed 2024-04-11T07:02:40Z
publishDate 2022-03-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS Computational Biology
spelling doaj.art-a8882e979b7d4715acc9a9a6eec8a8632022-12-22T04:38:38ZengPublic Library of Science (PLoS)PLoS Computational Biology1553-734X1553-73582022-03-01183e100993110.1371/journal.pcbi.1009931Determining clinically relevant features in cytometry data using persistent homology.Soham MukherjeeDarren WethingtonTamal K DeyJayajit DasCytometry experiments yield high-dimensional point cloud data that is difficult to interpret manually. Boolean gating techniques coupled with comparisons of relative abundances of cellular subsets is the current standard for cytometry data analysis. However, this approach is unable to capture more subtle topological features hidden in data, especially if those features are further masked by data transforms or significant batch effects or donor-to-donor variations in clinical data. We present that persistent homology, a mathematical structure that summarizes the topological features, can distinguish different sources of data, such as from groups of healthy donors or patients, effectively. Analysis of publicly available cytometry data describing non-naïve CD8+ T cells in COVID-19 patients and healthy controls shows that systematic structural differences exist between single cell protein expressions in COVID-19 patients and healthy controls. We identify proteins of interest by a decision-tree based classifier, sample points randomly and compute persistence diagrams from these sampled points. The resulting persistence diagrams identify regions in cytometry datasets of varying density and identify protruded structures such as 'elbows'. We compute Wasserstein distances between these persistence diagrams for random pairs of healthy controls and COVID-19 patients and find that systematic structural differences exist between COVID-19 patients and healthy controls in the expression data for T-bet, Eomes, and Ki-67. Further analysis shows that expression of T-bet and Eomes are significantly downregulated in COVID-19 patient non-naïve CD8+ T cells compared to healthy controls. This counter-intuitive finding may indicate that canonical effector CD8+ T cells are less prevalent in COVID-19 patients than healthy controls. This method is applicable to any cytometry dataset for discovering novel insights through topological data analysis which may be difficult to ascertain otherwise with a standard gating strategy or existing bioinformatic tools.https://doi.org/10.1371/journal.pcbi.1009931
spellingShingle Soham Mukherjee
Darren Wethington
Tamal K Dey
Jayajit Das
Determining clinically relevant features in cytometry data using persistent homology.
PLoS Computational Biology
title Determining clinically relevant features in cytometry data using persistent homology.
title_full Determining clinically relevant features in cytometry data using persistent homology.
title_fullStr Determining clinically relevant features in cytometry data using persistent homology.
title_full_unstemmed Determining clinically relevant features in cytometry data using persistent homology.
title_short Determining clinically relevant features in cytometry data using persistent homology.
title_sort determining clinically relevant features in cytometry data using persistent homology
url https://doi.org/10.1371/journal.pcbi.1009931
work_keys_str_mv AT sohammukherjee determiningclinicallyrelevantfeaturesincytometrydatausingpersistenthomology
AT darrenwethington determiningclinicallyrelevantfeaturesincytometrydatausingpersistenthomology
AT tamalkdey determiningclinicallyrelevantfeaturesincytometrydatausingpersistenthomology
AT jayajitdas determiningclinicallyrelevantfeaturesincytometrydatausingpersistenthomology