scispace - formally typeset
Open AccessPosted ContentDOI

A reference tissue atlas for the human kidney

Reads0
Chats0
TLDR
In this article, the authors describe the construction of an integrated reference tissue map of cells, pathways and genes using unaffected regions of nephrectomy tissues and undiseased human biopsies from 55 subjects.
Abstract
Kidney Precision Medicine Project (KPMP) is building a spatially-specified human tissue atlas at the single-cell resolution with molecular details of the kidney in health and disease. Here, we describe the construction of an integrated reference tissue map of cells, pathways and genes using unaffected regions of nephrectomy tissues and undiseased human biopsies from 55 subjects. We use single-cell and -nucleus transcriptomics, subsegmental laser microdissection bulk transcriptomics and proteomics, near-single-cell proteomics, 3-D nondestructive and CODEX imaging, and spatial metabolomics data to hierarchically identify genes, pathways and cells. Integrated data from these different technologies coherently describe cell types/subtypes within different nephron segments and interstitium. These spatial profiles identify cell-level functional organization of the kidney tissue as indicative of their physiological functions and map different cell subtypes to genes, proteins, metabolites and pathways. Comparison of transcellular sodium reabsorption along the nephron to levels of mRNAs encoding the different sodium transporter genes indicate that mRNA levels are largely congruent with physiological activity.This reference atlas provides an initial framework for molecular classification of kidney disease when multiple molecular mechanisms underlie convergent clinical phenotypes.

read more

Content maybe subject to copyright    Report

1
Towards Building a Smart Kidney Atlas: Network-based integration of multimodal
transcriptomic, proteomic, metabolomic and imaging data in the Kidney Precision
Medicine Project
Jens Hansen
1,*
, Rachel Sealfon
2,*
, Rajasree Menon
3,*
, Michael T. Eadon
4
, Blue B. Lake
5
,
Becky Steck
3
, Dejan Dobi
6
, Samir Parikh
7
, Tara K. Sidgel
6
, Theodore Alexandrov
8
, Andrew
Schroeder
6
, Edgar A. Otto
3
, Christopher R. Anderton
9,10
, Daria Barwinska
4
, Guanshi Zheng
10
,
Michael P. Rose
3
, John P. Shapiro
7
, Dusan Velickovic
9
, Annapurna Pamreddy
10
, Seth
Winfree
4
, Yongqun He
3
, Ian H. de Boer
11
, Jeffrey B. Hodgin
3
, Abhijit Nair
3
, Kumar Sharma
10
,
Minnie Sarwal
6
, Kun Zhang
5
, Jonathan Himmelfarb
11
, Zoltan Laszik
6
, Brad Rovin
7
, Pierre C.
Dagher
4
, John Cijiang He
1
, Tarek M. El-Achkar
4
, Sanjay Jain
12
, Olga G. Troyanskaya
2,#
,
Matthias Kretzler
3,#
, Ravi Iyengar
1,#
, Evren U. Azeloglu
1,#
for the Kidney Precision Medicine
Project Consortium
* Contributed equally, joint first authors
Affiliations:
1. Icahn School of Medicine at Mount Sinai, New York, New York
2. Princeton University, Princeton, New Jersey and Flatiron Institute, New York, New York
3. University of Michigan School of Medicine, Ann Arbor, Michigan
4. Indiana University School of Medicine, Indianapolis, Indiana
5. University of California San Diego, Jacobs School of Engineering, San Diego, California
6. University of California San Francisco School of Medicine, San Francisco, California
7. Ohio State University College of Medicine, Columbus, Ohio
8. European Molecular Biology Laboratory, Heidelberg, Germany
9. Pacific Northwest National Laboratory, Richland, Washington
10. UT-Health San Antonio School of Medicine, San Antonio, Texas
11. University of Washington, Schools of Medicine and Public Health, Seattle, Washington
12. Washington University in Saint Louis School of Medicine, St. Louis, Missouri
#
Corresponding Authors, joint senior authors:
Evren U. Azeloglu, Ph.D.
Assistant Professor of Medicine, Nephrology
Icahn School of Medicine at Mount Sinai, New York, NY
Email: evren.azeloglu@mssm.edu
Twitter: @azeloglu
Ravi Iyengar, Ph.D.
Dorothy H and Lewis H Rosenstiel Professor of Pharmacological Sciences
Icahn School of Medicine at Mount Sinai, New York, NY
Email: ravi.iyengar@mssm.edu
Matthias Kretzler, M.D.
Professor of Medicine, Nephrology
University of Michigan School of Medicine, Ann Arbor, MI
Email: kretzler@med.umich.edu
Olga Troyanskaya, Ph.D.
Professor of Computer Science
Princeton University, Princeton, NJ
Email: ogt@genomics.princeton.edu
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint

2
ABSTRACT
The Kidney Precision Medicine Project (KPMP) plans to construct a spatially specified
tissue atlas of the human kidney at a cellular resolution with near comprehensive molecular
details. The atlas will have maps of healthy, acute kidney injury and chronic kidney disease
tissues. To construct such maps, we integrate different data sets that profile mRNAs, proteins
and metabolites collected by five KPMP Tissue Interrogation Sites. Here, we describe a set of
hierarchical analytical methods to process, combine, and harmonize single-cell, single-nucleus
and subsegmental laser microdissection (LMD) transcriptomics with LMD and near single-cell
proteomics, 3-D nondestructive and immunofluorescence-based Codex imaging and spatial
metabolomics datasets. We use nephrectomy, healthy living donor and surveillance transplant
biopsy tissues to create a harmonized reference tissue map. Our results demonstrate that
different assays produce reliable and coherent identification of cell types and tissue
subsegments. They further show that the molecular profiles and pathways are partially
overlapping yet complementary for cell type-specific and subsegmental physiological
processes. Focusing on the proximal tubules, we find that our integrated systems biology-
based analyses identify different subtypes of tubular cells with potential for different levels of
lipid oxidation and energy generation. Integration of our omics data with pathways from the
literature, enables us to construct predictive computational models to develop a smart kidney
atlas. These integrated models can describe physiological capabilities of the tissues based on
the underlying cell types and pathways in health and disease.
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint

3
INTRODUCTION
The kidney is one of the most diverse organs in the human body in terms of its cellular
heterogeneity, and possibly second only to the brain in its spatial complexity. Accordingly,
decoding the functional and pathogenic mechanisms of kidney disease has been challenging;
as such, nephrology has consistently ranked behind all other subspecialties of medicine in
terms of the drug discovery pipeline
1
. Delineating the cell types and subtypes in different
regions of the kidney during health and disease will help identify the tissue-level, cellular and
subcellular pathways and processes involved in disease initiation and progression, and aid in
drug discovery.
The Kidney Precision Medicine Project (KPMP) is a consortium funded by the National
Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) that aims to ethically and
safely obtain kidney biopsies from participants with chronic kidney disease (CKD) or acute
kidney injury (AKI); create a reference kidney atlas; characterize disease subgroups to stratify
patients based on molecular features of disease; and identify critical cells, pathways, and
targets for novel therapies and preventive strategies. The KPMP features an expanding set of
complementary set of high throughput assays for molecular entities that span transcriptomic,
proteomic, metabolomic profiles and spatial/structural properties of kidney tissue. These
assays, described here for the five initially funded Tissue Interrogation Sites (TISes), will be
integrated to create a comprehensive knowledge environment for the human kidney. This
knowledge environment will be compiled by the KPMP Central Hub to serve as a foundation
for a spatially specified interactive smart tissue atlas that will include molecular and
physiological information on healthy and diseased states of all individual cell types within the
adult human kidney.
The KPMP envisions that harmonization and integration of different types of molecular data
from omics assays, combined with state-of-the-art pathological and clinical descriptors, will
allow us to classify different disease subtypes and states for diagnostic and therapeutic
purposes. Numerous groups have proposed the use of integrated multiomics analysis to
characterize disease phenotypes using tools that include Bayesian, correlative, network-based
and machine learning-based clustering algorithms
2-4
. The goals of these approaches include
prediction of clinical outcomes, identification of underlying disease mechanisms and
stratification of patients
5
. KPMP further envisions that the final integrated analytical
environment will serve as a knowledge base for the entire field that will empower a molecular
anchored outcome prediction and development of targeted treatments.
Here, we present an overview of KPMP’s strategies to harmonize and integrate multiple
data types through identification of subcellular pathways and functions that delineate cell-level
biochemical and physiological functions. Using reference kidney pilot tissue samples, we have
performed data harmonization and integration to investigate the complementarity of different
data types and develop a pipeline for the generation of tissue maps.
RESULTS
Outline of KPMP Data Types
In these analyses, there were four transcriptomic, two proteomic, one imaging-based, and
one spatial metabolomics tissue interrogation assays that consisted of 3 to 48 different
datasets obtained from 3 to 22 participants (Supplementary Table 1). These assays and their
detailed tissue pre-analytical, tissue processing, data acquisition and analytical data
processing pipelines are outlined in Figure 1. We also summarize the steps whereby the data
sets were integrated and harmonized in the upper right side of this descriptive map view of the
KPMP data integration paradigm.
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint

4
Pathway- and network-level integration of multiple molecular interrogation techniques
reveals cell- and tissue-specific biological processes that are critical for renal
physiology
To overcome the inherent challenges of multiomics integration and assay dependent
divergence, we employed dynamic enrichment analysis
6
and network mapping
7
. We
evaluated the convergence of subcellular processes (SCPs) and pathways that are over-
represented in different cell types or subsegments within the kidney (in comparison to the other
cell types or subsegments), using single cell RNASeq data from PREMIERE TIS (Michigan,
Princeton, Broad)
8
, single nucleus RNASeq data from UCSD/WU TIS
9
, Laser microdissected
(LMD) bulk RNASeq (Supplementary Table 2) and LMD proteomics (Supplementary Table 3)
from the OSU/IU TIS, Near Single Cell (NSC) proteomics from the UCSF TIS (Supplementary
Table 4) and spatial metabolomics from the UTHSA-PNNL-EMBL TIS (Supplementary Table
5A/B/C from 3 different participants).
Single-cell
8
and -nucleus
9
RNASeq analysis resulted in the grouping of multiple cells or
nuclei into clusters that were assigned to a particular cell type based on the expression of
essential genes. The top 300 most significantly differentially expressed genes (DEGs) and
proteins (DEPs) of each cluster or subsegment compared to all other clusters or subsegments
as well as the metabolites assigned to glomerular and non-glomerular kidney regions
(Supplementary Table 6) were subjected to enrichment analysis to create pathway maps
(Supplementary Table 7) for the three representative cell types contributing diverse function to
kidney physiology: proximal tubular epithelial cells (Figure 2A, Supplementary Figure 1A for
nonspecific pathways), podocytes (Supplementary Figure 1B) and principal cells of the
collecting ducts (Supplementary Figure 1C). The final maps revealed highly interrelated SCPs
that are intimately linked to the physiological function of the respective cell types. Furthermore,
these SCPs are highly overlapping between assays and datasets with up to 74% of them being
repeatedly enriched in two or more assays, confirming the inherent agreement among these
different assays. While the individual significant genes or gene products coming from multiple
assays were not necessarily the same, placement of these gene products into an
interconnected pathway map showed innate congruence between the assays. The key
subcellular processes (SCPs) for the different cell types differed significantly.
Cell-type specific SCP networks predict overlapping and complementary pathways that
accurately support each cell type’s whole cell function. Proximal tubule networks predict a high
metabolic activity and describe ion reabsorption and ion-triggered glucose reabsorption
pathways as well as ammonia metabolism and detoxification pathways (Figure 2A). The
predictions are in agreement with the energy intensive ion, glucose and other small molecule
reabsorption by the proximal tubule cells
10
and their predominant function in ammonium
excretion and renal drug clearance
11
. The identification of cellular iron homeostasis pathways
documents the iron storage capacity of proximal tubule cells
12
that among other functions,
also mitigates kidney damage during acute kidney injury
13
. Podocyte/glomerular networks
focus on cell-cell/cell-matrix adhesion, glomerular basement membrane/extracellular matrix
(ECM) and actin dynamics (Supplementary Figure 1B), all pathways fundamental for barrier
generation and consequently for glomerular filtration. Principal cell/collecting duct networks
concentrate on ion reabsorption (Supplementary Figure 1C), emphasizing the important role of
the collecting duct in fine-tuning these mechanisms, thereby regulating systemic electrolyte
and water balance.
These networks document that 13% (principal cells/collecting duct), 27% (proximal tubule
cells/tubulointerstitium) and 74% (podocytes/glomerulus) of all predicted SCPs were
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint

5
discovered by at least two different technologies. A closer investigation of the SCPs further
highlights that the overlap is even higher, if only the SCPs that describe cell type specific
functions are considered. Furthermore, the different datasets describe complementary
subfunctions of the same physiological processes. For example, both proteomic datasets of
the proximal tubule subsegments describe fatty acid transport via carnitine shuttling into the
mitochondrial matrix, where the enzymes for mitochondrial beta oxidation are localized (Figure
2A). The PREMIERE SC RNASeq dataset predicts carnitine biosynthesis, i.e. synthesis of the
central molecule of the carnitine shuttle.
Integration of pathways that were predicted based on the tubulointerstitial metabolites, such
as ‘Glycolysis and Gluconeogenesis’ and ‘D-Arginine and D-ornithine metabolism’
(Supplementary Figure 1D), into the Molecular Biology of the Cell Ontology (MBCO) SCP-
networks (Figure 1A) further underline the predicted high metabolic activity of the proximal
tubule cells. Glomerular metabolites enrich for pathways (Supplementary Figure 1C), such as
sphingolipid and arachidonic acid metabolism, that support cell-matrix/cell-cell adhesion and
gap junctions, respectively
14
. Dynamic enrichment analysis of both single-cell RNA-seq
datasets predicts the involvement of another metabolic pathway, i.e. retinol metabolism, in
podocyte function, in particular as a regulator of tight junctions (Supplementary Figure 1B).
Retinoic acid has a regulatory effect on tight junctions
15, 16
and plays a significant role in
mitigating podocyte apoptosis and dedifferentiation during podocyte injury
17
.
The enrichment results suggest that proximal tubular cells have the capacity to meet the
high energy demand by not only fueling the citric acid cycle via beta oxidation, but also via
glucose and glutamine catabolism. Nevertheless, beta oxidation is most consistently predicted,
in agreement with previous studies documenting lipid metabolism as the preferential energy
source in proximal tubule cells
18, 19
. Investigation of the pathway components of these SCPs
documents that the different omics technologies identify different components of these
pathways that integrate into a comprehensive description of the relevant biochemical pathways
(Figure 2B). Each technology contributes genes, proteins and metabolites for a fuller
description of the pathways than would be obtained by a single technology. Tubulointerstitial
metabolites, for example, contain glucose, cofactors of the pyruvate dehydrogenase complex
and multiple adenosine nucleotides/nucleosides (i.e. metabolites of the energy carrier ATP). In
agreement with the results of the pathway predictions, network mapping
7
revealed that cell-
type specific DEGs and DEPs lie within the same area of the human interactome
(Supplementary Figure 1E), indicative of close functional relationships.
In parallel, we identified modules in a kidney-specific functional network using the top
ranked 300 marker genes and proteins across all datatypes in order to detect sets of cell-type
specific, functionally related genes
20, 21
. The module detection algorithm finds groups of genes
that form tightly connected communities within a kidney-specific functional network, which is
constructed using a data-driven approach from gene-gene relationships across thousands of
experimental assays. After module detection, gene enrichment analysis is performed within
each module to understand the key functions of the genes in each module. As with dynamic
enrichment analysis, the modules display clear cell-type specific functional enrichments
(Supplementary Table 8). For example, the network of proximal tubule marker genes includes
modules enriched in anion transport and cellular response to metal ions (Figure 2C), the
network of podocyte marker genes includes modules enriched in glomerulus development and
cell-cell adhesion (Supplementary Figure 1F), and the network of principal cell marker genes
includes modules enriched in sodium ion transport (Supplementary Figure 1G)
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 24, 2020. ; https://doi.org/10.1101/2020.07.23.216507doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI

How to Get Started with Single Cell RNA Sequencing Data Analysis.

TL;DR: In this paper, a review of the analysis pipelines used in the field of single-cell analysis is presented, along with an overview of challenges and the most commonly used analytical tools.
Journal ArticleDOI

Perspectives in systems nephrology.

TL;DR: In this paper, a holistic approach aims to identify the molecular basis of CKD subtypes as well as individual determinants of disease manifestation in a given patient, which will lead to improved prognostic and predictive diagnostics and the discovery of novel molecular disease-specific therapies.
References
More filters
Journal ArticleDOI

Kidney physiology and susceptibility to acute kidney injury: implications for renoprotection.

TL;DR: In this paper, the authors discuss why specific kidney regions are at particular risk of injury and how this information might help to delineate novel routes for mitigating injury and avoiding permanent damage, and suggest that the physiological heterogeneity of the kidney should be taken into account when exploring novel renoprotective strategies, such as improvement of kidney tissue oxygenation, stimulation of hypoxia signalling pathways and modulation of cellular energy metabolism.
Journal ArticleDOI

Novel renal amino acid transporters

TL;DR: Reabsorption of amino acids, similar to that of glucose, is a major task of the proximal kidney tubule and cationic amino acids and some other neutral amino acids leave the cell basolaterally via heterodimeric obligatory exchangers.
Journal ArticleDOI

Apical Transporters for Neutral Amino Acids: Physiology and Pathophysiology

TL;DR: Recent advances in the molecular identification of apical neutral amino acid transporters has shed a light on the molecular basis of Hartnup disorder and iminoglycinuria.
Journal ArticleDOI

Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets.

TL;DR: In this article, the authors profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution and reveal key cell type-specific transcription factors and major gene-regulatory circuits for kidney cells.
Related Papers (5)
Frequently Asked Questions (15)
Q1. What have the authors contributed in "Towards building a smart kidney atlas: network-based integration of multimodal transcriptomic, proteomic, metabolomic and imaging data in the kidney precision medicine project" ?

Towards Building a Smart Kidney Atlas: Network-based integration of multimodal transcriptomic, proteomic, metabolomic and imaging data in the Kidney Precision Medicine Project Jens Hansen, Rachel Sealfon, Rajasree Menon, Michael T. Eadon, Blue B. Lake, Becky Steck, Dejan Dobi, Samir Parikh, Tara K. Sidgel, Theodore Alexandrov, Andrew Schroeder, Edgar A. Otto, Christopher R. Anderton, Daria Barwinska, Guanshi Zheng, Michael P. Rose, John P. Shapiro, Dusan Velickovic, Annapurna Pamreddy, Seth Winfree, Yongqun He, Ian H. de Boer, Jeffrey B. Hodgin, Abhijit Nair, Kumar Sharma, Minnie Sarwal, Kun Zhang, Jonathan Himmelfarb, Zoltan Laszik, Brad Rovin, Pierre C. Dagher, John Cijiang He, Tarek M. El-Achkar, Sanjay Jain, Olga G. Troyanskaya, Matthias Kretzler, Ravi Iyengar, Evren U. Azeloglu for the Kidney Precision Medicine Project Consortium 

Their approach is amendable to future computational modeling studies that can further improve the proposed tissue atlas. In addition to the integrated analytics presented here, the KPMP is also building a community-based Kidney Tissue Atlas Ontology ( KTAO ), which will systematically integrate different types information ( such as clinical, pathological, cell and molecular ) into a logically defined tissue atlas, which can then be further utilized to support various applications 34. 

‘SCTransform’ was used for data normalization and scaling (based on top 2,000 features), followed by principal component analysis. 

Decrease in fatty acid oxidation, resulting in a loss of ATP generation, has been shown to be a significant contributor to tubulointerstitial fibrosis 19. 

On average 12 and 15 libraries (~3,100 and 3,835 nuclei) allowed reidentification of seven of the top 10 predicted podocyte and proximal tubule MBCO SCPs, respectively, while 21 libraries (~5,462 nuclei) were sufficient to reidentify five of the topwas not certified by peer review) is the author/funder. 

Subcellular localization of each gene was identified using the jensenlab human compartment database based on a jensenlab confidence of at least four (i.e. 80% of maximum confidence in the database) 28. 

Tubulointerstitial metabolites, for example, contain glucose, cofactors of the pyruvate dehydrogenase complex and multiple adenosine nucleotides/nucleosides (i.e. metabolites of the energy carrier ATP). 

Their results indicate that for a consistent detection of podocytes (i.e. in more than 95% of all down sampled datasets with the same library counts), at least 16 (~11,727 cells) or 7 libraries (1,837 nuclei) are needed if subjected to single-cell RNASeq (Figure 4A) or single-nucleus RNASeq (Figure 4B), respectively. 

Top 300 differentially expressed genes (DEGs) and proteins (DEPs) predicted by each assay for each analyzed cell type/tissue subsegment. 

Notice that the top seven predictions based on dynamic enrichment analysis can contain more than seven SCPs, since each prediction is either a single SCP or a unique combination of two or three SCPs. 

For the LMD proteomics dataset, six to eight samples were sufficient to reproduce the results obtained for the full datasets with only minor variations in the correlation of identified DEGs (Figure 4C) and SCPs (Supplementary Figure 2E) or SCP rankings (Figure 4C). 

An idealized integration scenario would combine these assays synergistically such that they could complement the shortcomings of each other, improve quality control metrics across technologies, and increase rigor and reproducibility of the overall study. 

the authors determined how many SCPs have to be considered in a down-sampled analysis to re-identify at least 70% (or 50%) of the top 10 or seven predictions obtained from standard or dynamic enrichment analysis with the full dataset, respectively. 

Principal cell/collecting duct networks concentrate on ion reabsorption (Supplementary Figure 1C), emphasizing the important role of the collecting duct in fine-tuning these mechanisms, thereby regulating systemic electrolyte and water balance. 

To compute the Pearson correlation between the gene expression profiles of cells and LCM segments, the gene profiles were restricted to genes shared between the two datasets and showing variable expression in the single-cell dataset and correlations were computed between the logarithm of the mean ratio vector for each LCM segment and the scaled expression profile of each cell in the single cell dataset.