scispace - formally typeset
Search or ask a question
Posted ContentDOI

Targeted single-cell RNA-seq identifies minority cell types of kidney distal nephron that regulate blood pressure and calcium balance

19 Jul 2020-bioRxiv (Cold Spring Harbor Laboratory)-
TL;DR: This work coupled a FACS-enrichment protocol with single-cell RNA-seq analysis to profile the transcriptomes of 9099 cells from the nephron region adjacent to the macula densa and creates a web-based data resource for these cells.
Abstract: A major objective in modern biology is generation of comprehensive atlases of various organs identifying all cell types and their expressed genes. In kidney, extensive data exists for proximal tubule and collecting duct cells, but not for non-abundant intermediate epithelial cell types. Here, we coupled a FACS-enrichment protocol with single-cell RNA-seq analysis to profile the transcriptomes of 9099 cells from the nephron region adjacent to the macula densa. Clusters containing Slc12a3+/Pvalb+ and Slc12a3+/Pvalb- cells were identified as DCT1 and DCT2 cells. The DCT1 cells appear to be heterogeneously associated with variable expression of Slc8a1, Calb1, and Ckb among other mRNAs. No DCT2-specific transcripts were found. The analysis also identified two distinct cell types in the Slc12a1+ portion of Henle’s loop as well as Nos1+/Avpr1a+ macula densa cells. Thus, we identify unexpected cell diversity in the intermediate region of the nephron and create a web-based data resource for these cells.

Summary (2 min read)

INTRODUCTION

  • The mammalian kidneys play a crucial role in regulation of body fluid composition and blood pressure.
  • BioRxiv preprint 4 cells have been indicative of variable expression along the DCT with detectable levels only in the distal-most DCT14, 15.
  • The progressive development and improvement of RNA-seq for comprehensive quantification of gene expression, especially small-sample RNA-seq in microdissected renal tubules17, and more recent single-cell RNA-seq techniques in kidney18, 19, 20, 21, 22, 23, 24, 25, have provided a deep analysis of gene expression in the kidney epithelial cells and have broadened their insights into renal cell identities and functions.

RESULTS

  • To provide appropriate context for single-cell RNA-seq (scRNA-seq) analysis of cells in nephron region from cortical thick ascending limb of Henle (CTAL) to the distal convoluted tubule (DCT) to the connecting tubule (CNT) in mouse, the authors began by mapping transcriptomes in microdissected CTAL, DCT, and CNT tubule segments.
  • To characterize gene expression in CTAL, DCT, and CNT plus all other renal tubule segments, the authors performed renal tubule microdissection under a standard stereomicroscope (Fig. 1b) followed by small-sample RNA-seq (Supplementary Data 1).
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.
  • In DCT, some transcripts with no prior known functional roles in the DCT were identified.
  • The transcriptomes of each cell cluster are provided in Supplementary Data 7.

Discussion

  • A complete understanding of the cellular composition and transcriptional profiles of the cells in DCT has been lacking.
  • The authors observed that DCT1 cells are heterogeneous.
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.
  • A striking observation from the present study is the identification of two CTAL subtypes based on gene expression that might correspond to the morphologically identified cell types, namely cells corresponding to the C5 cluster (Foxq1+Cldn10-Ptger3-) and the C6 cluster (Foxq1Cldn10+Ptger3+).
  • In summary, systematic analyses of cellular composition and gene expression provide insights into kidney functions and diseases.

Methods

  • Adult mouse kidney cell isolation and single-cell RNA-seq.
  • The final cell pellet was resuspended in 30ml FACS buffer (perfusion buffer with 0.05% BSA) and then passed sequentially through 100 µm, 70 µm and 40 µm cell strainers (VWR) to obtain a suspension of isolated cells (single-cell suspension).
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.
  • The source data underlying Figs. 2a,d, 3a-b, 4a and Supplementary Figs.

Acknowledgments

  • The work was primarily funded by the Division of Intramural Research, National Heart, Lung, and Blood Institute (project ZIA-HL001285 and ZIA-HL006129, M.A.K.).
  • Confocal images were taken in the NHLBI Confocal Microscopy Core Facility (Christian Combs, Director).
  • Cell sorting was performed in the NHLBI Flow Cytometry Core (J. Philip McCoy, Director).
  • Tissue sections were prepared in the NHBLI Pathology Facility (Zu-Xi Yu, Director). was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.

Author contributions

  • Was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.

Figure Legends

  • Overview of renal tubule cell nomenclature and experimental design.
  • Fig. 2. Gene expression pattern among mouse renal tubule segments.
  • The copyright holder for this preprint (whichthis version posted July 19, 2020.
  • The same color codes were used for exons in the splice graph and UCSC Genes Track.
  • Heterogeneity of distal convoluted tubule cells revealed by single-cell RNA-seq.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
Targeted single-cell RNA-seq identifies minority cell types of kidney distal nephron that
regulate blood pressure and calcium balance
Lihe Chen
1
, Chun-Lin Chou
1
, and Mark A. Knepper
1*
1
Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood
Institute, National Institutes of Health, Bethesda, Maryland
Running title: Single-cell RNA-seq Reveals Transcriptomes of DCT1, DCT2, and Subtypes of CTAL
Cells
*Correspondence and Lead Contact: Mark A. Knepper, MD, PhD, Senior Investigator, Division of
Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892-1603
Email: knepperm@nhlbi.nih.gov
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 19, 2020. ; https://doi.org/10.1101/2020.07.19.209627doi: bioRxiv preprint

2
ABSTRACT
A major objective in modern biology is generation of comprehensive atlases of various
organs identifying all cell types and their expressed genes. In kidney, extensive data exists for
proximal tubule and collecting duct cells, but not for non-abundant intermediate epithelial cell
types. Here, we coupled a FACS-enrichment protocol with single-cell RNA-seq analysis to profile
the transcriptomes of 9099 cells from the nephron region adjacent to the macula densa.
Clusters containing Slc12a3
+
/Pvalb
+
and Slc12a3
+
/Pvalb
-
cells were identified as DCT1 and DCT2
cells. The DCT1 cells appear to be heterogeneously associated with variable expression of
Slc8a1, Calb1, and Ckb among other mRNAs. No DCT2-specific transcripts were found. The
analysis also identified two distinct cell types in the Slc12a1
+
portion of Henle’s loop as well as
Nos1
+
/Avpr1a
+
macula densa cells. Thus, we identify unexpected cell diversity in the
intermediate region of the nephron and create a web-based data resource for these cells.
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 19, 2020. ; https://doi.org/10.1101/2020.07.19.209627doi: bioRxiv preprint

3
INTRODUCTION
The mammalian kidneys play a crucial role in regulation of body fluid composition and
blood pressure. These functions are largely achieved by control of sodium reabsorption across
the epithelia of renal tubules
1, 2
. There are at least 14 different renal tubule segments, each
with characteristic cell types with distinct gene expression profiles and function
3, 4
. Of particular
importance is the distal convoluted tubule (DCT), a short segment that mediates fine regulation
of sodium ion transport
5
. The apical component of sodium transport across the DCT occurs via
the Na
+
-Cl
-
cotransporter NCC (Slc12a3), which is exclusively expressed in DCT cells and the
chief target of thiazide diuretics
6
, a staple in the treatment of many forms of arterial
hypertension. The DCT is believed to be heterogeneous and is separated into at least two
subsegments, DCT1 and DCT2
7, 8, 9
. Both express Slc12a3. DCT1 but not DCT2 expresses the
Ca
2+
-binding protein, parvalbumin (Pvalb). DCT2, in contrast to DCT1, expresses the epithelial
sodium channel (ENaC)
9
. More recent observations indicate that the DCT2 also expresses the
Ca
2+
transporter, Trpv5
10
, and has been proposed to be a site of transepithelial Ca
2+
transport
9,
11
. However, this DCT classification is largely defined based on immunocytochemical results. It
remains unclear how many cell types are in DCT and whether DCT1 and DCT2 are distinct
segments or represent two ends of a continuum.
NaCl transport across the DCT was originally thought to be affected in part by binding of
circulating aldosterone to the mineralocorticoid receptor (MR) in DCT cells
12, 13
. Aldosterone-
MR signaling requires an enzyme 11-β-hydroxysteroid dehydrogenase 2 (Hsd11b2) to
deactivate the more abundant circulating cortisol, which has an equal binding affinity to MR.
However, immunocytochemical and in situ hybridization methods to localize Hsd11b2 in DCT
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 19, 2020. ; https://doi.org/10.1101/2020.07.19.209627doi: bioRxiv preprint

4
cells have been indicative of variable expression along the DCT with detectable levels only in
the distal-most DCT
14, 15
. These findings raised doubts about a possible direct role of
aldosterone in the early DCT, and whether the Hsd11b2 positive DCT corresponds to DCT2.
Recent studies further suggest that Slc12a3 (NCC) abundance is regulated by aldosterone only
indirectly via its propensity to cause hypokalemia
5, 16
. This process is thought to be controlled
by multiple receptors, protein kinases (Wnk1, Wnk4, and SPAK), ubiquitin ligase components
(Nedd4l, Klhl3, Cul3), potassium channels, and chloride channels
5, 16
. However, a systematic
mapping of these elements in DCT is currently not available.
The progressive development and improvement of RNA-seq for comprehensive
quantification of gene expression, especially small-sample RNA-seq in microdissected renal
tubules
17
, and more recent single-cell RNA-seq techniques in kidney
18, 19, 20, 21, 22, 23, 24, 25
, have
provided a deep analysis of gene expression in the kidney epithelial cells and have broadened
our insights into renal cell identities and functions. The tubule microdissection method
26
,
however, is unable to effectively isolate the DCT1 and DCT2 due to the overall shortness of DCT
and the lack of a distinct transition point between DCT1 and DCT2. Despite great progress of
comprehensive single-cell RNA-seq in the kidney (‘shotgun’ scRNA-seq), it devotes most of the
sequencing reads to more abundant proximal tubule cells and non-epithelial cells, leading to
analysis of only limited numbers of minority epithelial cell types. Thus, the heterogeneity of cell
types like DCT cells as well as various cell types in the loop of Henle (for example, macula densa
and cortical thick ascending limb cells) are not completely resolved. A solution to this dilemma
is to enrich the target cells before analysis as previously introduced for scRNA-seq analysis of
collecting duct cell types
19
.
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 19, 2020. ; https://doi.org/10.1101/2020.07.19.209627doi: bioRxiv preprint

5
Here, we describe an enrichment protocol prior to single-cell RNA-seq analysis and
specially focused on the mouse nephron region spanning from the cortical thick ascending limb
of Henle (CTAL) to the DCT. In parallel, we use small-sample RNA-seq to characterize the gene
expression in all 14 renal tubules microdissected from mouse kidneys. We also provide web-
based resources that allow users to explore and download data for future studies.
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 19, 2020. ; https://doi.org/10.1101/2020.07.19.209627doi: bioRxiv preprint

Citations
More filters
01 Jan 1998
TL;DR: In this paper, an affinity-purified peptide-directed antibody was used to detect a 165-kDa band in membrane fractions from the renal cortex but not from renal medulla, which indicated that the distal convoluted tubule is an important site of action of the mineralocorticoid aldosterone.
Abstract: Although the collecting duct is regarded as the primary site at which mineralocorticoids regulate renal sodium transport in the kidney, recent evidence points to the distal convoluted tubule as a possible site of mineralocorticoid action. To investigate whether mineralocorticoids regulate the expression of the thiazide-sensitive Na-Cl cotransporter (TSC), the chief apical sodium entry pathway of distal con- voluted tubule cells, we prepared an affinity-purified, peptide- directed antibody to TSC. On immunoblots, the antibody recognized a prominent 165-kDa band in membrane fractions from the renal cortex but not from the renal medulla. Immu- nof luorescence immunocytochemistry showed TSC labeling only in distal convoluted tubule cells. Semiquantitative im- munoblotting studies demonstrated a large increase in TSC expression in the renal cortex of rats on a low-NaCl diet (207 6 21% of control diet). Immunof luorescence localization in tissue sections confirmed the strong increase in TSC expres- sion. Treatment of rats for 10 days with a continuous subcu- taneous infusion of aldosterone also increased TSC expression (380 6 58% of controls). Furthermore, 7-day treatment of rats with an orally administered mineralocorticoid, f ludrocorti- sone, increased TSC expression (656 6 114% of controls). We conclude that the distal convoluted tubule is an important site of action of the mineralocorticoid aldosterone, which strongly up-regulates the expression of TSC.

12 citations

Posted ContentDOI
24 Jul 2020-bioRxiv
TL;DR: A set of hierarchical analytical methods to process, combine, and harmonize single-cell, single-nucleus and subsegmental laser microdissection (LMD) transcriptomics and 3-D nondestructive and immunofluorescence-based Codex imaging and spatial metabolomics datasets are described.
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 biologybased 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.

5 citations

Posted ContentDOI
15 Sep 2021-bioRxiv
TL;DR: 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.

3 citations

References
More filters
Journal ArticleDOI
TL;DR: The Spliced Transcripts Alignment to a Reference (STAR) software based on a previously undescribed RNA-seq alignment algorithm that uses sequential maximum mappable seed search in uncompressed suffix arrays followed by seed clustering and stitching procedure outperforms other aligners by a factor of >50 in mapping speed.
Abstract: Motivation Accurate alignment of high-throughput RNA-seq data is a challenging and yet unsolved problem because of the non-contiguous transcript structure, relatively short read lengths and constantly increasing throughput of the sequencing technologies. Currently available RNA-seq aligners suffer from high mapping error rates, low mapping speed, read length limitation and mapping biases. Results To align our large (>80 billon reads) ENCODE Transcriptome RNA-seq dataset, we developed the Spliced Transcripts Alignment to a Reference (STAR) software based on a previously undescribed RNA-seq alignment algorithm that uses sequential maximum mappable seed search in uncompressed suffix arrays followed by seed clustering and stitching procedure. STAR outperforms other aligners by a factor of >50 in mapping speed, aligning to the human genome 550 million 2 × 76 bp paired-end reads per hour on a modest 12-core server, while at the same time improving alignment sensitivity and precision. In addition to unbiased de novo detection of canonical junctions, STAR can discover non-canonical splices and chimeric (fusion) transcripts, and is also capable of mapping full-length RNA sequences. Using Roche 454 sequencing of reverse transcription polymerase chain reaction amplicons, we experimentally validated 1960 novel intergenic splice junctions with an 80-90% success rate, corroborating the high precision of the STAR mapping strategy. Availability and implementation STAR is implemented as a standalone C++ code. STAR is free open source software distributed under GPLv3 license and can be downloaded from http://code.google.com/p/rna-star/.

30,684 citations

Journal ArticleDOI
TL;DR: EdgeR as mentioned in this paper is a Bioconductor software package for examining differential expression of replicated count data, which uses an overdispersed Poisson model to account for both biological and technical variability and empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference.
Abstract: Summary: It is expected that emerging digital gene expression (DGE) technologies will overtake microarray technologies in the near future for many functional genomics applications. One of the fundamental data analysis tasks, especially for gene expression studies, involves determining whether there is evidence that counts for a transcript or exon are significantly different across experimental conditions. edgeR is a Bioconductor software package for examining differential expression of replicated count data. An overdispersed Poisson model is used to account for both biological and technical variability. Empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference. The methodology can be used even with the most minimal levels of replication, provided at least one phenotype or experimental condition is replicated. The software may have other applications beyond sequencing data, such as proteome peptide count data. Availability: The package is freely available under the LGPL licence from the Bioconductor web site (http://bioconductor.org).

29,413 citations

Journal ArticleDOI
TL;DR: It is shown that accurate gene-level abundance estimates are best obtained with large numbers of short single-end reads, and estimates of the relative frequencies of isoforms within single genes may be improved through the use of paired- end reads, depending on the number of possible splice forms for each gene.
Abstract: RNA-Seq is revolutionizing the way transcript abundances are measured. A key challenge in transcript quantification from RNA-Seq data is the handling of reads that map to multiple genes or isoforms. This issue is particularly important for quantification with de novo transcriptome assemblies in the absence of sequenced genomes, as it is difficult to determine which transcripts are isoforms of the same gene. A second significant issue is the design of RNA-Seq experiments, in terms of the number of reads, read length, and whether reads come from one or both ends of cDNA fragments. We present RSEM, an user-friendly software package for quantifying gene and isoform abundances from single-end or paired-end RNA-Seq data. RSEM outputs abundance estimates, 95% credibility intervals, and visualization files and can also simulate RNA-Seq data. In contrast to other existing tools, the software does not require a reference genome. Thus, in combination with a de novo transcriptome assembler, RSEM enables accurate transcript quantification for species without sequenced genomes. On simulated and real data sets, RSEM has superior or comparable performance to quantification methods that rely on a reference genome. Taking advantage of RSEM's ability to effectively use ambiguously-mapping reads, we show that accurate gene-level abundance estimates are best obtained with large numbers of short single-end reads. On the other hand, estimates of the relative frequencies of isoforms within single genes may be improved through the use of paired-end reads, depending on the number of possible splice forms for each gene. RSEM is an accurate and user-friendly software tool for quantifying transcript abundances from RNA-Seq data. As it does not rely on the existence of a reference genome, it is particularly useful for quantification with de novo transcriptome assemblies. In addition, RSEM has enabled valuable guidance for cost-efficient design of quantification experiments with RNA-Seq, which is currently relatively expensive.

14,524 citations

Journal ArticleDOI
TL;DR: An overview of DAMP-sensing receptors is provided, the crosstalk between these receptors is discussed, and the cross-regulation of these receptors and their ligands are discussed.
Abstract: The innate immune system has the capacity to detect ‘non-self’ molecules derived from pathogens, known as pathogen-associated molecular patterns, via pattern recognition receptors. In addition, an increasing number of endogenous host-derived molecules, termed damage-associated molecular patterns (DAMPs), have been found to be sensed by various innate immune receptors. The recognition of DAMPs, which are produced or released by damaged and dying cells, promotes sterile inflammation, which is important for tissue repair and regeneration, but can also lead to the development of numerous inflammatory diseases, such as metabolic disorders, neurodegenerative diseases, autoimmune diseases and cancer. Here we examine recent discoveries concerning the roles of DAMP-sensing receptors in sterile inflammation and in diseases resulting from dysregulated sterile inflammation, and then discuss insights into the cross-regulation of these receptors and their ligands. Host-derived molecules, the so-called damage-associated molecular patterns (DAMPs), can induce sterile inflammation. This Review provides an overview of DAMP-sensing receptors, discusses the crosstalk between these receptors and explores their role in disease.

762 citations

Journal ArticleDOI
18 May 2018-Science
TL;DR: It is inferred that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type, and that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell.
Abstract: Our understanding of kidney disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ’s multiple homeostatic functions. To help fill this knowledge gap, we characterized 57,979 cells from healthy mouse kidneys by using unbiased single-cell RNA sequencing. On the basis of gene expression patterns, we infer that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type. We also found that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell. Computational cell trajectory analysis and in vivo lineage tracing revealed that intercalated cells and principal cells undergo transitions mediated by the Notch signaling pathway. In mouse and human kidney disease, these transitions were shifted toward a principal cell fate and were associated with metabolic acidosis.

751 citations

Related Papers (5)