You are on page 1of 11

Am J Physiol Gastrointest Liver Physiol 320: G439–G449, 2021.

First published January 27, 2021; doi:10.1152/ajpgi.00397.2020

RESEARCH ARTICLE

Inflammation, Immunity, Fibrosis, and Infection

Noninvasive stratification of nonalcoholic fatty liver disease by whole


transcriptome cell-free mRNA characterization
Naga Chalasani,1 Shusuke Toden,2 John J. Sninsky,2 Richard P. Rava,2 Jerome V. Braun,2 Samer Gawrieh,1
Jiali Zhuang,2 Michael Nerenberg,2 Stephen R. Quake,3 and Tara Maddala2
1
Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis,
Indiana; 2Molecular Stethoscope, San Diego, California; and 3Departments of Bioengineering and Applied Physics, Stanford
University and Chan Zuckerberg Biohub, Stanford, California

Abstract
Hepatic fibrosis stage is the most important determinant of outcomes in patients with nonalcoholic fatty liver disease (NAFLD).
There is an urgent need for noninvasive tests that can accurately stage fibrosis and determine efficacy of interventions. Here,
we describe a novel cell-free (cf)-mRNA sequencing approach that can accurately and reproducibly profile low levels of circulat-
ing mRNAs and evaluate the feasibility of developing a cf-mRNA-based NAFLD fibrosis classifier. Using separate discovery and
validation cohorts with biopsy-confirmed NAFLD (n = 176 and 59, respectively) and healthy subjects (n = 23), we performed se-
rum cf-mRNA RNA-Seq profiling. Differential expression analysis identified 2,498 dysregulated genes between patients with
NAFLD and healthy subjects and 134 fibrosis-associated genes in patients with NAFLD. Comparison between cf-mRNA and liver
tissue transcripts revealed significant overlap of fibrosis-associated genes and pathways indicating that the circulating cf-mRNA
transcriptome reflects molecular changes in the livers of patients with NAFLD. In particular, metabolic and immune pathways re-
flective of known underlying steatosis and inflammation were highly dysregulated in the cf-mRNA profile of patients with
advanced fibrosis. Finally, we used an elastic net ordinal logistic model to develop a classifier that predicts clinically significant fi-
brosis (F2–F4). In an independent cohort, the cf-mRNA classifier was able to identify 50% of patients with at least 90% probabil-
ity of clinically significant fibrosis. We demonstrate a novel and robust cf-mRNA-based RNA-Seq platform for noninvasive
identification of diverse hepatic molecular disruptions and for fibrosis staging with promising potential for clinical trials and clini-
cal practice.
NEW & NOTEWORTHY This work is the first study, to our knowledge, to utilize circulating cell-free mRNA sequencing to de-
velop an NAFLD diagnostic classifier.

cell-free mRNA; classification biomarker; NASH; nonalcoholic fatty liver disease; nonalcoholic steatohepatitis

INTRODUCTION Although liver biopsy remains the preferred reference


method for NASH and NAFLD diagnosis (5), biopsies are
Nonalcoholic fatty liver disease (NAFLD) is the most com- invasive, expensive, subjective due to pathology review,
mon liver disease in the world, affecting one-quarter of the and prone to sampling error. In addition, although numer-
global population (1, 2). The more common form of NAFLD ous pharmacotherapies are in development, none is yet
is nonalcoholic fatty liver (NAFL) or simple steatosis, approved for NASH, and many have failed in recent trials
whereas a subset of patients with NAFLD develop the pro- (6–9). Intensive lifestyle intervention and bariatric surgery
gressive form, nonalcoholic steatohepatitis (NASH). NASH is have demonstrated reductions in steatosis and fibrosis but
characterized by steatosis, lobular inflammation, and hepa- would benefit from noninvasive efficacy metrics beyond
tocyte ballooning and is associated with increasing levels of just weight loss, an imperfect correlate of liver steatosis
liver fibrosis, which can subsequently progress to liver cir- (10–13). Therefore, there is a critical need for accurate non-
rhosis, liver failure requiring transplant, and hepatocellular invasive tests for NASH and advanced fibrosis.
carcinoma (3). Identification of patients with NASH and Currently, there are two distinct approaches for noninva-
advanced hepatic fibrosis is critical due to their greater risk sive assessment of liver fibrosis, a “physical” approach based
for developing complications of chronic liver disease (4). on the assessment of liver stiffness using elastography

Correspondence: T. Maddala (tmaddala@molecularstethoscope.com); N. Chalasani (nchalasa@iu.edu).


Submitted 30 October 2020 / Revised 17 December 2020 / Accepted 8 January 2021

http://www.ajpgi.org 0193-1857/21 Copyright © 2021 the American Physiological Society G439


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

techniques and a “biological” approach based on the quanti- intercept the intercellular communication signals and in-
fication of biomarkers in serum samples (14). For imaging- terrogate the molecular changes associated with NALFD
based approaches, vibration-controlled transient elastogra- progression.
phy (VCTE) and magnetic resonance elastography are the
most widely used clinical tools (15–17). As for quantitative bi- MATERIALS AND METHODS
ological approaches, blood-based tests such as fibrosis 4
(FIB-4) index, which accounts for aspartate aminotransfer- Patients and Sample Information
ase and alanine aminotransferase levels, the platelet count,
Samples from two NAFLD cohorts (one retrospective, “dis-
and age to give a fibrosis index, is used commonly in the
covery cohort” and one prospective, “validation cohort”)
clinics (7, 18). Furthermore, several circulating protein-based
and one prospective healthy cohort were evaluated
tests including the enhanced liver fibrosis (ELF) test as well
(Supplemental Table S1 and S2; all supplemental material is
as NIS4, which combines protein markers and circulating
available at https://doi.org/10.5061/dryad.1jwstqjt1). A total
microRNA, miR-34a, levels, are also available (15, 19).
of 188 serum samples were retrospectively randomly selected
Although these diagnostic approaches have been useful to
from the Indiana University School of Medicine biobank (IU)
estimate the severity of fibrosis in patients with NAFLD,
to obtain a balanced representation of liver fibrosis (F0–F4),
there are limited data on their utility in monitoring fibrosis
of which 176 were evaluable and had matched liver biopsy
progression/regression (20).
evaluated by central pathological reading. These 176 samples
Over the past decade, an increasing research focus on “liq-
were allocated as the “discovery cohort.” A total of 89 serum
uid biopsy” has resulted in the development of several circu-
samples were collected prospectively from IU and the biore-
lating nucleic acid-based diagnostic tests for multiple
pository at the University of Florida (UF), using a standar-
diseases (21). Accordingly, various types of circulating
dized collection protocol. From UF, 59 were evaluable due to
nucleic acids, including cell-free DNA (cf-DNA), methylated
having matched liver biopsy evaluated by a local pathologist.
cf-DNAs, and noncoding RNAs, especially microRNAs, have
Inclusion criteria for the both retrospective and prospective
been examined as potential noninvasive biomarker candi-
cohort specified suspected NAFLD and NASH diagnosed via
dates for NAFLD (22–25). In recent gene-expression profiling
biopsy, which was to be performed within 30 days of blood
studies of liver tissue acquired from patients with NAFLD/
collection. Patients were also required to fast for 8 h before
NASH, gene-expression signatures for patients with NAFLD/
blood draw. Patients with other forms of liver disease (hepa-
NASH and transcriptionally regulated pathways associated
titis B or C, alcoholic hepatitis, etc.), patients taking drugs
with increased disease activity and fibrosis were discovered
known to cause hepatic steatosis, and pregnant patients
(26). Therefore, messenger RNAs appear to be promising bio-
were excluded. In brief, blood samples were collected in BD
marker candidates for NAFLD/NASH diagnosis and monitor-
Vacutainer clotting tubes (BD No. 367820) and processed
ing. Although quantification of cell-free messenger RNAs
within 2 h after the blood draw. All samples were centrifuged
(cf-mRNAs) was considered challenging due to their low
abundance in the circulation, we and others have developed at 1,900 g for 10 min; then, serum was separated into new
next-generation sequencing (NGS)-based platforms and tubes and stored at 80 C. All 59 biopsy-confirmed prospec-
demonstrated that circulating cf-mRNA profiling may be tive NAFLD samples were used as the “validation cohort.” In
used for diagnosis and monitoring of multiple diseases (27– addition, serum samples from 23 control healthy individuals
29). Studies have shown that rather than cf-mRNA reflecting were prospectively collected from the San Diego Blood Bank,
debris from cellular apoptosis or necrosis as with cfDNA, CA, using the same standardized protocol. Written informed
these selective actively released circulating nucleic acids consent was obtained from all patients, and the study was
provide lines of intercellular communication to inform prox- approved by the institutional review boards of all the partici-
imal and distal cells of ongoing active transcriptional proc- pating institutions. Clinical data used in the study were
esses (30, 31). Furthermore, beyond a reflection of ongoing obtained from the individual institutions in March 2018 and
cellular processes, cf-mRNA has been demonstrated to be were collected and verified by each of the participating
biologically functional such that cells that take up these institutions.
nucleic acids have altered transcription (32). Histology and Biochemical Tests
Here, using a robust NGS platform developed to measure
transcript levels in circulation, we sequenced the cf-mRNA All biopsies were routinely stained with hematoxylin
transcriptomes from the serum of 247 well-characterized and eosin and Masson’s trichrome. Liver pathologists at
patients with NAFLD and 23 healthy control individuals. We the individual institute scored stained sections using the
first established the potential of our technology to differenti- NASH Clinical Research Network scoring system (35). A
ate NAFLD from healthy controls to identify dysregulated fasting blood sample was obtained at the time of biopsy,
pathological processes. Next, we identified cf-mRNA tran- and routine biochemical tests were performed using
scriptomic signatures associated with the continuum of liver standard methods and assays. Biochemical tests included
fibrosis, many of which coincide with previously known bio- aminotransferase (ALT) and aspartate aminotransferase
logical processes of disease (33, 34). Using these differen- (AST), and additional blood samples were drawn for se-
tially expressed genes, we developed a cf-mRNA-based rum processing.
classifier to stratify patients according to the severity of liver
Library Preparation and Whole Transcriptome RNA-Seq
fibrosis and assessed performance in an independent cohort
of patients with NAFLD. Our study demonstrates the poten- RNA was extracted using the QIAamp Circulating Nucleic
tial of using cf-mRNA profiling as a “liquid-biopsy” to Acid kit (Qiagen) from up to 1 mL of serum and eluted in a

G440 AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

16 mL volume. Then, 1 mL of extracted RNA was analyzed on Cf-mRNA Transcriptome Decomposition Using
an Agilent RNA 6000 Pico chip (Agilent Technologies) for Nonnegative Matrix Factorization
presence of observable RNA traces. In total, 5 mL of the
extracted RNA was then converted into a sequencing library Nonnegative matrix factorization (NMF) was performed to
as described previously (30). Qualitative and quantitative decompose normalized gene-expression profiles from cf-
analyses of the NGS library preparation process were con- mRNA into 12 components. In NMF decomposition, genes
ducted using a chip-based electrophoresis, and libraries sharing similar expression patterns across samples are
were quantified using a qPCR-based quantification kit grouped together in an unsupervised manner. NMF decom-
(Roche, Cat. No. KK4824). Sequencing was performed using position was performed using the scikit-learn Python
the Illumina NextSeq500 platform (Illumina Inc.), using machine-learning library. Genes with >40% loading attrib-
paired-end sequencing, 75-cycle sequencing. For all sequenc- utable to a particular component were considered enriched
ing data, we obtained a median of 8.7 million pass-filter reads in the component. For each component, we selected genes
per sample (range: 8.1–16.2 million reads). Base-calling was enriched in the component and examined associated path-
performed on an Illumina BaseSpace platform (Illumina ways and biological processes using Gene Ontology via
Inc.), using the FASTQ Generation Application. For sequenc- Metascape (37). To robustly perform Gene Ontology analysis,
ing data analysis, adaptor sequences were removed, and low- we selected clusters with at least 200 genes as a criterion for
quality bases were trimmed using cutadapt (v1.11). Reads further analysis. We determined the title of the cluster by
shorter than 15 base-pairs were excluded from subsequent preferentially using the top category identified in the Gene
analysis. Read sequences greater than 15 base-pairs were Ontology biological processes. If the terminology provided
compared with the human reference genome GRCh38 using by the biological process category was nonspecific, we eval-
STAR (v2.5.2b) with GENCODE v24 gene models. Duplicated uated the top term identified in the pathway analysis and
reads were removed using the samtools (v1.3.1) rmdup com- named the cluster according to the pathway term.
mand. Gene-expression levels were calculated from dedupli-
Bioinformatics Analysis/Classifier
cated BAM files using RSEM (v1.3.0).
In each sample for each gene, we took the arithmetic aver-
Multiplex qPCR age of the TPMs of the replicates. Because of sample size
limitations in the discovery cohort, fibrosis stage was aggre-
cDNA was generated by primer annealing 3 mL of RNA with gated to three categories: F0/F1, F2, and F3/F4. We applied
10 mM dNTPs (Thermo Fisher Scientific, 10297018) and ran- an ordinal logistic regression model with an elastic net pen-
dom hexamers (IDT) at 60 C for 5 min, followed by reverse alty (equal weighting to ridge and lasso) to classify fibrosis
transcription using the SuperScript IV Reverse Transcriptase
stage. Predictiveness curves were generated for the valida-
kit (Invitrogen, 18090200) according to the manufacturer’s
tion cohort. These curves depict the probability of clinically
instruction. Subsequently, cDNA was amplified using
significant fibrosis (F2–F4) on the y-axis and the percentile
Platinum Taq DNA polymerase (Invitrogen, 10966034)
of the biomarker score on the x-axis.
and preamplification primers (Supplemental Table S3).
The cDNA samples were then treated with Exonucleus I
(Thermo Fischer Scientific, EN0582) to remove primers
used for the nested PCR reactions according to the manu- RESULTS
facturer’s instruction. Multiplex qPCR was conducted
Cohort Descriptions
with the Fluidigm BioMark HD system (Fluidigm) using
96.96 Dynamic array IFC (Fluidigm) with SsoFast Evaluable samples from three cohorts of patients were
EvaGreen Supermixes (Bio-Rad, 1725200) with target gene included in analyses: 176 NASH/NAFL patient samples from
primers (primer sequences are listed in Supplemental a retrospective cohort (discovery cohort), 59 NASH/NAFL
Table S3). patient samples from a prospective cohort (validation
cohort), and 23 healthy control samples collected prospec-
Differential Expression Analysis and Correlation tively from the San Diego Blood Bank (Fig. 1 and
Differential expression (DE) analysis was implemented Supplemental Table S1). The full fibrosis spectrum of disease
with DESeq2 (v1.12.4) (36) using read counts as input. Genes was observed in both NAFLD cohorts (Supplementary Table
with fewer than 200 total reads across the entire cohort were S2). The age and BMI distributions were reflective of the gen-
excluded from subsequent analysis. Technical replicates eral NAFLD population and were similar in both NAFLD
were averaged before the DE analysis, which applies a nega- cohorts. Of note, the proportion of females was higher in the
tive binomial model with raw read counts as the dependent discovery cohort compared with the validation and healthy
variable. For NAFLD versus healthy control analysis, the in- control cohorts (Supplemental Table S1). Furthermore, the
dependent variables were NAFLD/healthy and sex. For anal- proportion of females increased with higher fibrosis in the
ysis of fibrosis, fibrosis was treated as a continuous discovery cohort (Supplemental Table S2). The distributions
variable. The Benjamini–Hochberg correction was used to of some demographics and baseline factors were imbalanced
correct for multiple testing and to obtain adjusted P val- across cohorts and/or across fibrosis stage. Common comor-
ues in differential expression analysis. Gene Ontology bidities within this cohort were examined. In this study, 19%
and Reactome Pathway enrichment analyses were con- of patients with NAFLD were confirmed diabetics, and the
ducted using Metascape (37) using Homo sapiens as the median BMI among NAFLD patients was 34 kg/m2, which is
input and analysis species. considered obese.

AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org G441


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

RNA and subsequently used for multiplex qPCR profiling of


Cohort 96 genes. RNA-Seq TPM values inversely correlated with the
NAFL/NASH Enrolled (n = 247) qPCR cycle threshold (CT) value (Pearson’s correlation, r =
• Discovery (Retrospective samples, n = 188)
0.86), indicating a high degree of concordance between cf-
• Validation (Prospective samples, n = 59)
mRNA-seq and qPCR platforms (Fig. 2D). Collectively, these
data highlight the technical robustness of the cf-mRNA
Fibrosis Retrospective Prospective Total
RNA-Seq assay in individuals with NAFLD.
F0 53 9 62
F1 53 14 67
F2 38 11 49 Identification of Transcriptomic Signatures in
F3 24 9 33 Circulation Associated with NAFLD Compared with
F4 20 16 36 Healthy Controls
To evaluate dysregulated cf-mRNA transcripts that are
Excluded: Samples with distinct to patients with NAFLD, we conducted differential
hemolysis gene expression analysis between NAFLD (discovery cohort)
( n = 12 retrospective) and healthy controls. Applying the DEseq algorithm (using a
negative binomial model) to raw read counts (36), and
Analyzable data (n = 235)
adjusting for sex, we identified 2,498 differentially expressed
• Discovery (Retrospective samples, n = 176)
• Validation (Prospective samples, n = 59) genes [false discovery rate (FDR) < 0.05, Fig. 3A and
Supplemental File S1]. Of these genes, 1,527 genes were up-
Fibrosis Retrospective Prospective Total regulated and 971 genes were downregulated. Subsequently,
F0 51 9 60 using these dysregulated genes, we applied Gene Ontology
F1 49 14 63 and Reactome pathway analyses to identify pathways that
F2 35 11 46 are dysregulated in the cf-mRNA transcriptomes of patients
F3 22 9 31 with NAFLD. Gene Ontology analysis revealed dysregulation
F4 19 16 35 of immune system process and metabolic process as well
Figure 1. Consort diagram. Summary of patient samples that were col- as change in cellular component organization or biogene-
lected initially and subsequent samples that were used for the analysis. sis; all common processes that are associated with fibrosis
Samples that were excluded from the study are also summarized. (33) (Fig. 3B, Supplemental Fig. S2A, and Supplemental
File 1). Interestingly, we identified “localization” as the
most dysregulated pathway for both upregulated and
Technical Performance of cf-mRNA NGS Assay in
downregulated genes. Previous studies have indicated that
Human Serum Specimens
liver fibrosis dysregulates genes that are associated with
We have previously demonstrated the potential of cf- cellular localization (42, 43). Considering that dysregula-
mRNA as a platform for diagnosis and monitoring disease tion of genes that are associated with “localization” can be
status in patients with hematological cancers (30) and in overexpressed or inhibited in the liver, we speculate that
Alzheimer’s disease (38). We examined the robustness of the this GO term was identified in both groups. Similarly,
cf-mRNA platform in liver disease specifically, by evaluating Reactome pathway analysis identified dysregulation of fibro-
the technical reproducibility of libraries generated from cf- sis-associated pathways, including the adaptive immune sys-
RNA in all the serum samples in this study. For most sam- tem, Rho GTPase signaling, and angiogenesis (Supplemental
ples (239/247, 97%) multiple serum aliquots were available Fig. 2B and Supplemental File 1).
and technical replicates were analyzed. Whole transcriptome Next, to examine the overall biology of the cf-mRNA
comparison [transcripts per million (TPM)  10] of technical transcriptome in patients with NAFLD, we applied unsu-
replicates showed high correlation indicative of robust tech- pervised nonnegative matrix factorization (NMF) (27)
nical reproducibility (Pearson’s correlation  0.94 for 95% of decomposition of cf-mRNA transcriptomes using NAFLD
NAFLD samples) (Fig. 2A). In addition, the high correlation samples from the discovery cohort. Subsequently, we iden-
between observed versus expected number of extracellular tified 12 distinct gene clusters (Fig. 3C and Supplemental
RNA consensus consortium (ERCC) molecules demonstrated File S2). Of 12 clusters, we focused specifically on six clus-
high quantification accuracy (Fig. 2B and Supplemental Fig. ters where the number of genes in the cluster was greater
S1A), 95% of samples with Pearson’s correlation r  0.93) than 200 (Supplemental Fig. 2C). We then used Gene
(39). A median of 7,120 transcripts with  10 TPM were iden- Ontology to identify key biological processes and pathways
tified per sample (Fig. 2C), highlighting the diversity of the that are associated with individual clusters (Fig. 3D and
circulating mRNA transcripts quantified by the assay. To fur- Supplemental Fig. S2C). Functional analyses revealed iden-
ther validate the robustness of the cf-mRNA RNA-Seq assay, tification of the following clusters: cellular component or-
we compared the levels of circulating transcripts assessed by ganization, immune systems 1 and 2, metabolic process,
cf-mRNA RNA-Seq with qPCR. We used multiplex qPCR hemostasis, and cellular localization (Supplemental Fig.
(Fluidigm BioMark) to assess the expression of 96 gene tar- S2C). Interestingly, several clusters such as metabolic pro-
gets known to be expressed in healthy and NAFLD liver tis- cess, cellular component organization, and immune sys-
sue at different levels (40, 41) (Fig. 2D, Supplemental Table tems are well-recognized biological processes that are
3). Twelve plasma samples from the discovery cohort, sepa- associated with NAFLD, indicating that molecular altera-
rate aliquots to those used for RNA-Seq, were used to extract tions in the liver might be reflected in the circulation.

G442 AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

A B
50
Sample number

40 20

30

Count
20 10

10 Figure 2. Cell-free (cf)-mRNA-based high-


throughput sequencing is an accurate, ro-
0 0 bust, and reproducible approach for charac-
0.88 0.92 0.96 1.0 0.900 0.925 0.950 0.975 terizing serum transcriptome. A: histogram
representing correlation between the cf-
Pearson Correlation Coefficient (r) Pearson’s correlation coefficient (r)
mRNA transcriptomes of technical replicates
Independent using Pearson’s correlation analysis (n =
RNA extraction 239). B: histogram compiling r values of
C D RNA NGS 96 genes was expected extracellular RNA consensus con-
Multiplex assessed in 12 sortium (ERCC) transcripts vs. observed
RNA qPCR
patients level of ERCC from each sample (n = 239).
C: histogram of number of transcripts
30 10000 detected (transcripts per million, TPM 10)
r = -0.81
per sample (n = 239). D: schematics for com-
1000 P < 0.0001
parison between next-generation sequenc-
Sample number

RNA-Seq (log TPM)

20 ing assay and qPCR (top). Correlation


100 between sequencing TPM against qPCR Ct
value (96 genes in total, n = 12) (bottom).
10
10
1

0.1
0
6000 7000 8000 0.01
5 10 15 20 25 30 35 40
Number of protein-coding genes
detected (TPM ≥ 10) qPCR (CT value)

Collectively, our data indicate that the cf-mRNA profile can metabolic processes and immune system processes, both
be used to noninvasively identify molecular alterations that well-known processes that are commonly dysregulated in
are specific to patients with NAFLD, and the gene-expres- patients with NAFLD (Fig. 4B). In addition, we used
sion profile of patients with NAFLD may reflect the molecu- Reactome pathway analysis to examine biological path-
lar characteristics of the disease. ways that are linked to cf-mRNA fibrosis-associated genes
(Supplemental Fig. 3B). The Reactome pathway analysis
Identification of Transcriptomic Signatures in identified pathways that are known to be associated with
Circulation Associated with Fibrosis Stage within the fibrosis and NAFLD, including notch pathways and lipid
NAFLD Cohort metabolism (44, 45). We then used publicly available liver
NAFLD-related transcriptomic changes in serum cf- tissue RNA sequencing data that were generated from
mRNA were evaluated in the discovery NAFLD cohort sam- patients with NAFLD and liver fibrosis (34) and compared
ples to identify markers of fibrosis. Applying the DESeq algo- genes that are associated with fibrosis between hepatic tis-
rithm to raw read counts and treating fibrosis as a sue and cf-mRNA. The tissue RNA-Seq data were gener-
continuous variable, we applied a less stringent threshold of ated from liver biopsy samples from 72 patients with
FDR < 0.1 due to the more challenging problem of identify- NAFLD, and ordinal regression was applied to identify
ing differentially expressed genes within the NAFLD patient genes that are associated with fibrosis stages (34). A total
cohort. We identified 134 differentially expressed genes of 4,010 genes were identified using FDR < 0.05 as the cut-
(FDR < 0.1, Fig. 4A, Supplemental File S2). The majority (79, off criterion. The comparison between differentially
59%) of genes were upregulated, whereas 55 (41%) genes expressed genes between tissues and cf-mRNA resulted in
were downregulated (Fig. 4A). The terms “upregulated” and identification of 43 common genes (32% of fibrosis associ-
“downregulated” are used to describe changes in the number ated cf-mRNA genes) (Fig. 4C). Furthermore, we evaluated
of RNA molecules in the circulation of higher fibrosis the common pathways that are dysregulated in fibrosis
patients. Next, we used Gene Ontology and Reactome path- among both tissue and cf-mRNA (Fig. 4C). We identified
way analysis algorithms to evaluate the functional roles and that of the 21 tissue and 17 cf-mRNA Gene Ontology biolog-
biological processes reflected by these differentially ical pathways identified, 15 pathways were common
expressed genes (Fig. 4B and Supplemental Fig. S3, A and between liver tissues and cf-mRNA (Fig. 4C). Collectively,
B). Using Gene Ontology enrichment analysis, cf-mRNA our data suggest that dysregulated genes in the cf-mRNA
genes that are associated with fibrosis stages were associ- reflect those of tissues and frequently display common
ated with biological processes of NAFLD, including dysregulated pathways.

AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org G443


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

A B Most significant -log(p-value)


biological processes 0 20 40 60 80
Controls vs NAFLD localization
negative regulation of biological process

Upregulated
metabolic process
10.0 APOC1
multi-organism process
APOB cellular component organization or biogenesis
-log10(adjusted p-value)

HPD
response to stimulus
7.5 APOC3 CRTAC1 cellular process
immune system process
regulation of biological process
5.0 developmental process
LMBRD2

1 0 20 40 60 80
2.5
localization
Adj. p < 0.05 immune system process

Downregulated
0.0 response to stimulus
biological regulation
-2 0 2 multicellular organismal process
2
-log (fold change) biological adhesion
cellular component organization or biogenesis
cellular process
signaling
locomotion

C D Clusters
70
60

-log(p-value) 50
40
Hemostasis
30
20
10
0
cellular component organization or biogenesis

biological regulation

immune system process

metabolic process

localization

immune system process


Immune process 1
biological processes
Most significant

Metabolic process
Immune process 2 0.4
0.2
Cellular localization
0.0
-0.2
Cellular component -0.4
organization

Figure 3. Cell-free (cf)-mRNA profile shows distinct profile for patients with nonalcoholic fatty liver disease (NAFLD) compared with control subjects. A:
volcano plot depicting the cf-mRNA genes that are differentially expressed between patients with NAFLD and healthy controls (n = 176 and 23, respec-
tively, n = number of subjects). Significantly dysregulated genes are denoted in red, false discovery rate (FDR) < 0.05 was used as the cutoff criterion. B:
most significant biological pathways identified using upregulated (top) and downregulated genes (bottom) identified in A as inputs (Gene Ontology). C:
heat map showing nonnegative matrix factorization (NMF) gene clusters within patients with NAFLD. D: top Gene Ontology biological process categories
associated with each cluster.

Although many patients with NASH have advanced liver with in those with NAFL (Fig. 4D). We identified 167 differen-
fibrosis, NASH is a distinct histological entity that is diag- tially expressed genes between NASH and NAFL (FDR < 0.10,
nosed based on pattern recognition of steatosis, inflamma- Fig. 4D, Supplemental File 3). Whereas 88 (53%) genes were
tion, and ballooning. We, therefore, examined cf-mRNA upregulated, 79 (47%) genes were downregulated (Fig. 4D).
genes that are dysregulated in patients with NASH compared Pathway analyses of these dysregulated genes revealed that

G444 AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

A Fibrosis associated genes


B C Overlapping genes
5 -log10(p-value)
Most significant Tissue Cf-mRNA
FSCN1
biological processes 0 1 2 3 4 5 6
4 metabolic process 3967 43 91
-log10(adjusted p-value)

AKR1B10
PIGR
cellular component organization or biogenesis
multicellular organismal process Adj. p = 0.1
3 AZU1
BPI signaling Adj. p = 0.05
FCN3 immune system process
2
SLC2A1 AKAP12 TCIM
negative regulation of biological process Common biological
biological adhesion processes
developmental process
1 Adj. p < 0.1
Tissue Cf-mRNA
response to stimulus
biological regulation
6 15 2
0
-0.5 0 0.5 1
-log2(fold change)

D NASH vs. NAFL E F


6 -log10(p-value)
Most significant
biological processes 0 1 2 3 4 5 6 7 8
Overlapping genes
5 FCN3 metabolic process
-log10(adjusted p-value)

response to stimulus
4 Fibrosis NASH
developmental process
biological regulation
98 36 131
3 GLOD4 localization
TRPC1 MAN1A1 ADRM1
NACC2 IKZF3 regulation of biological process
2 FSCN1
cellular process
positive regulation of biological process
1 Adj. p < 0.1
negative regulation of biological process
detoxification
0
-2 -1 0 1 2 3
-log2(fold change)
Figure 4. Cell-free (cf)-mRNA profiling reveals key biological pathways and processes associated with advanced fibrosis. A: volcano plot depicting the
differential expression analysis associated with fibrosis in cf-mRNA (n = 176). Significantly dysregulated genes are denoted in red, false discovery rate
(FDR) < 0.10 was used as the cutoff criterion. B: most significantly enriched pathways identified using genes significantly dysregulated with fibrosis
stages (Gene Ontology). C: overlap of genes (top) and pathways (bottom) between tissue and cf-mRNA fibrosis stage-associated genes [tissue data
obtained from Hoang et al. (34)]. Pathway analysis was conducted using Gene Ontology (biological processes). D: volcano plot depicting the differential
expression analysis between nonalcoholic steatohepatitis (NASH) and NAFL in cf-mRNA (132 NASH vs. 44 NAFL). Significantly dysregulated genes are
denoted in green, FDR < 0.10 was used as the cutoff criterion. E: most significantly enriched pathways identified using genes significantly dysregulated
between NASH and NAFLD (Gene Ontology). F: overlap of differentially dysregulated genes between fibrosis and NASH vs. NAFLD comparison.

metabolic pathways were prominent (Fig. 4E and Supplemen- modeled fibrosis as an ordinal variable (F0/F1, F2, F3/F4)
tal Fig. 3C) for both Gene Ontology and Reactome (Fig. 4E and and applied an elastic net penalty. The resulting biomarker
Supplemental Fig. 3, C and D). In addition, we compared dys- score was applied to the independent prospective NAFLD
regulated genes between fibrosis and NASH versus NAFL com- cohort. Rather than reporting AUC, sensitivity, and specific-
parison. Interestingly, only 36 genes that were dysregulated in ity, which assume a clinically actionable risk threshold and
both analyses overlapped, indicating that the molecular profile are dependent on the cohort in which these parameters are
of NASH substantially differs from that of advanced fibrosis estimated, we display the predicted probability of having
and would require a separate set of genes to effectively identify clinically significant fibrosis (F2 to F4) on the y-axis as a
patients with NASH (Fig. 4F). function of the percentile of the biomarker score on
the x-axis (Fig. 5A). This predictiveness curve shows the
range and distribution of estimated risk levels associated
Stratification of NAFLD Fibrosis Stages
with the model when it is applied to the population from
Since biopsy-measured fibrosis is an imperfect gold stand- which the cohort was drawn (46). Although the predictive-
ard, we developed a classifier of fibrosis stage that yields the ness curve relates to classification performance, the curve
patient’s estimated probability of having F2–F4 fibrosis. To also displays essential information about risk that is not dis-
assess the feasibility of distinguishing fibrosis stages for played by the receiver operating characteristic curve and
detection, disease management, and therapeutic trials, we highlights the ability of the biomarker score to tease apart
developed this classifier based on the discovery cohort. We risk from the average risk in the cohort (40% in this cohort).

AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org G445


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

A Fibrosis (F2/F3/F4) B Advanced fibrosis (F3/F4)

1.00 1.00

Probability of advanced fibrosis (F3/F4)


Probability of fibrosis (F2/F3/F4)

0.75 0.75

0.50 0.50

0.25 0.25

0.00 0.00
0.00 0.25 0.50 0.75 1.00 0.00 0.25 0.50 0.75 1.00
Percentile of linear predictor Percentile of linear predictor

Fibrosis categories
F0/F1 F2 F3/F4

Figure 5. Performance of a cell-free (cf)-mRNA fibrosis classifier. A: predicted probability of clinically significant fibrosis (F2, F3, and F4) in the validation
set. B: predicted probability of advanced fibrosis (F3 and F4) in the validation set.

Therefore, the predictiveness curve reflects both risk model- to diagnose and monitor the disease status of NAFLD, as
ing and population performance approach, providing a more well as to identify novel therapeutic targets. Blood-based
complete and comprehensive analysis than an evaluation of noninvasive assays may potentially overcome many of the
classifier performance. The predictiveness curve depicted in limitations intrinsic to liver biopsy and enable better disease
Fig. 5A shows 25% of patients, the majority of whom are la- screening, diagnosis, and monitoring (50). In the present
beled F0/1, with less than 25% probability of having F2–F4 study, we utilized cohorts with biopsy-confirmed fibrosis
disease. The figure also shows 40% of patients, the majority covering a broad spectrum of disease. Subsequently, we
of whom are labeled F2–F4, with higher than 90% probabil- identified cf-mRNA markers of NAFLD and NAFLD severity
ity of having F2–F4 disease. Similarly, we developed a classi- that are common to those observed in liver tissue of patients
fier for advanced fibrosis (F3/F4). Thirty-four percent of with NAFLD (34). We used two cohorts to demonstrate that
patients were classified with less than 25% probability of molecular alterations associated with liver fibrosis are
having advanced fibrosis, a majority of whom were F0/1/2; reflected in the circulation, and these transcriptional changes
36% of patients had higher than 90% probability of having may be used to develop classifiers to assess the severity and
advanced fibrosis, a majority of whom were F3–F4 (Fig. 5B). perhaps ongoing diverse pathological changes with interven-
Collectively, these data show that cf-mRNA transcriptome tion of liver fibrosis. We acknowledge that the present study
can be used to develop effective classifiers for liver fibrosis. is a proof-of-concept study and has limitations in its design
that may have introduced selection bias as well as unmeas-
DISCUSSION ured confounders. Further analytical and clinical develop-
ment and validation studies in the intended-use population
NAFLD is a disorder that affects a quarter of the world- are needed as a next step to developing a robust clinical-
wide population, resulting in a considerable global health grade NASH fibrosis diagnostic.
and economic burden (47). Although liver biopsy is currently Over the past decade, blood-based molecular biomarkers
the reference method for NAFLD characterization, this inva- have been identified as key candidates for the development of
sive approach has several key limitations. The biopsy sam- clinically relevant noninvasive disease biomarkers. Accord-
ples are equivalent to only one fifty-thousandth of the liver ingly, several circulating DNA-based biomarkers have been
volume and may not reflect the pathological status of the developed for prenatal diagnostics, transplant rejection, and
entire liver. Pathological grading of the biopsy samples cancer monitoring (51–56). Recent gene-expression profiling of
is subject to inter- and intraobserver variation (48, 49). liver tissues obtained from patients with NAFLD/NASH
Furthermore, liver biopsies are expensive, are invasive, and revealed unique gene-expression alterations for patients with
carry the risk, though infrequent, of serious complications, NAFLD compared with those of healthy individuals (26). For
thereby making them unsuitable for disease monitoring. For this reason, we believe that the signals observed from liver
these reasons, there is an urgent need for noninvasive tools reflect the expression of active processes and are reflective of

G446 AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

disease. In the present study, we demonstrated the utility of cir-


culating transcriptomic cf-mRNA profiling to evaluate molecu-
ACKNOWLEDGMENTS
lar alterations associated with patients with NAFLD. Although We thank Teresa Wright and Guillermo Elias for conceptual dis-
RNA-Seq-based cf-mRNA profiling has been used previously in cussions and critical reading of the manuscript; Neeraj Salathia,
other diseases (27–29), this is the first study, to our knowledge, Arkaitz Ibarra, John Aballi, Alex Acosta, Lucy Guo, Vera Huang,
Amy P Karns, Julianna R Parks, and Yue Zhao for technical input
to utilize the cf-mRNA RNA-Seq technology platform to com- and assistance; Kayla Gelow and Emily R Smith for sample collec-
prehensively examine the transcriptional alterations in patients tion and data management; Brooke McMillen Patz and the Clinical
with NAFLD and to evaluate the association of cf-mRNA pro- and Translational Sciences Institute (CTSI) Biorepository group at
files with severity of liver diseases. We showed that cf-mRNA Indiana University, and Erin Kelly and the CTSI Biorepository
profiling reflected that of previously performed liver tissue group at University of Florida, for sourcing patient samples; and
sequencing data and identified several pathways that were asso- Ruben Rodrigues and Brian Read at the Development Research
ciated with key biological processes that are linked to NAFLD, Services for assistance with sourcing samples from the San Diego
including lipid metabolism and extracellular matrix dysregula- Blood Bank.
tion. Our results demonstrate that cf-mRNA profiling can poten-
tially be used beyond simple disease diagnosis and fibrosis DISCLOSURES
assessment in liver disease by utilizing the genes and pathways S.T., J.J.S., R.P.R., J.V.B., J.Z., M.N., and T.M. are current or past
that are dysregulated in liver for therapeutic target identifica- employees at Molecular Stethoscope, Inc. S.T., J.Z., and M.N. are
tion, patient selection, and the monitoring of efficacy of phar- named as inventors in patent applications related to the technolo-
macotherapy, lifestyle intervention, and bariatric surgery. gies used in this manuscript. S.T., J.Z., and M.N. are named as
With numerous drugs currently being developed for inventors in pending patent applications related to the technolo-
NAFLD and a need to inform the encouragement of lifestyle gies used in this manuscript filed by Molecular Stethoscope
intervention or bariatric surgery, it is critical to identify the Inc. [WO2020092646A1 filed on October 30, 2019 (J.Z., M.N.),
WO2020087037A2 filed on October 25, 2019 (S.T., J.Z., M.N.),
stage of disease (57, 58). Although most current clinical trials
and WO2019060369A1 filed on September 18, 2018 (M.N.)]. S.R.
utilize a liver biopsy to identify a suitable patient population, Q. is a founder of Molecular Stethoscope, Inc., and a member of
the invasive nature of biopsy limits its utility as a broader its scientific advisory board. N.C. has ongoing consulting agree-
patient-screening tool for clinical trials (50), patient monitor- ments with Abbvie, Madrigal, Foresite, Zydus, ObsEva, and
ing during trials, or for monitoring intervention efficacy. The Galectin and research support from DSM, Exact Sciences, Zydus,
fibrosis-4 index (FIB4), NAFLD fibrosis score (NFS), NIS4, and and Intercept. At the time of this work, J.V.B. had ongoing consult-
vibration-controlled transient elastography (VCTE) are cur- ing agreements from Exact Sciences, CareDX, and Grail. These
rently available noninvasive tests to predict the presence and outside interests are not directly or significantly related to this pa-
severity of hepatic fibrosis in NAFLD (16, 17, 19, 50, 59). per. At the time of this work, T.M. had ongoing consulting agree-
ments and/or other compensation from CareDX, Delfi, Grail, and
However, these tests are based on simple variables such as Lexent Bio. These outside interests are not directly or significantly
age, liver enzymes, platelets, and liver physical stiffness, related to this paper. S.G. consults for TransMedics and Pfizer and
which unlike our cf-mRNA classifier are unlikely to reflect the receives research grant support from Zydus, Galmed and Viking.
real-time dynamics of NAFLD pathophysiology or hepatic None of these interests is related to this work. These outside
transcriptome changes in response to therapeutic interven- interests are not directly or significantly related to this paper. All
tions for NAFLD. We utilized whole transcriptome cf-mRNA authors declare that they have no additional competing interests.
profiling to develop a classifier for fibrosis. The classifier was No external funding was used for this study.
able to identify a significant proportion of patients at either
very high or low probability of having clinically significant fi- AUTHOR CONTRIBUTIONS
brosis (F2–F4) in the validation cohort. Since the classifier N.C., S.T., M.N., and T.M. conceived and designed research;
includes multiple gene transcripts, it is more likely to reflect N.C., S.T., J.J.S., R.P.R., J.V.B., J.Z., and T.M. analyzed data; N.C.,
the multiple underlying pathologies known to be involved in S.T., J.J.S., R.P.R., J.V.B., S.G., J.Z., M.N., S.R.Q. and T.M. inter-
this chronic complex disease. Because an expression-based preted results of experiments; S.T., J.V.B., and J.Z. prepared fig-
assay could be used to evaluate multiple aspects of the disease ures; N.C., S.T., J.Z., and T.M. drafted manuscript; N.C., S.T., J.J.S.,
and of patient response over time, our study demonstrates R.P.R., J.V.B., S.G., J.Z., M.N., S.R.Q., and T.M. edited and revised
the potential utility of cf-mRNA as a promising alternative to manuscript; N.C., S.T., J.J.S., R.P.R., J.V.B., S.G., J.Z., M.N., S.R.Q.,
liver biopsies, imaging, and conventional blood tests. and T.M. approved final version of manuscript.

DATA AVAILABILITY REFERENCES


Sequencing data generated in this study was deposited in 1. Danford CJ, Sanchez JE, Corey KE. Managing the burden of non-
Sequence Read Archive (SRA) under accession number NASH NAFLD. Curr Hepatol Rep 16: 326–334, 2017. doi:10.1007/
PRJNA701722 (https://www.ncbi.nlm.nih.gov/sra/PRJNA701722). s11901-017-0371-9.
All data needed to evaluate the conclusions in the paper are 2. Ludwig J, Viggiano TR, McGill DB, Oh BJ. Nonalcoholic steatohe-
present in the paper and/or the supplementary materials. patitis: mayo clinic experiences with a hitherto unnamed disease.
Mayo Clin Proc 55: 434–438, 1980.
3. Brunt EM. Pathology of nonalcoholic fatty liver disease. Nat Rev
SUPPLEMENTAL DATA Gastroenterol Hepatol 7: 195–203, 2010. doi:10.1038/nrgastro.
2010.21.
Supplemental Figures S1–3 and Tables S1–3: https://doi.org/10. 4. Caste  ra L, Foucher J, Bernard PH, Carvalho F, Allaix D,
5061/dryad.1jwstqjt1) Merrouche W, Couzigou P, de Le dinghen V. Pitfalls of liver

AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org G447


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

stiffness measurement: a 5-year prospective study of 13,369 17. Tapper EB, Castera L, Afdhal NH. FibroScan (vibration-controlled
examinations. Hepatology 51: 828–835, 2010. doi:10.1002/ transient elastography): where does it stand in the United States
hep.23425. practice. Clin Gastroenterol Hepatol 13: 27–36, 2015. doi:10.1016/j.
5. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, cgh.2014.04.039.
Harrison SA, Brunt EM, Sanyal AJ. The diagnosis and management 18. Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ;
of nonalcoholic fatty liver disease: practice guidance from the Nash Clinical Research Network. Comparison of noninvasive
American Association for the Study of Liver Diseases. Hepatology markers of fibrosis in patients with nonalcoholic fatty liver disease.
67: 328–357, 2018. doi:10.1002/hep.29367. Clin Gastroenterol Hepatol 7: 1104–1112, 2009. doi:10.1016/j.
6. Friedman SL, Ratziu V, Harrison SA, Abdelmalek MF, Aithal GP, cgh.2009.05.033.
Caballeria J, Francque S, Farrell G, Kowdley KV, Craxi A, Simon K, 19. Harrison SA, Ratziu V, Boursier J, Francque S, Bedossa P, Majd Z,
Fischer L, Melchor-Khan L, Vest J, Wiens BL, Vig P, Seyedkazemi Cordonnier G, Sudrik Fb Darteil R, Liebe R, Magnanensi J, Hajji Y,
S, Goodman Z, Wong VW, Loomba R, Tacke F, Sanyal A, Lefebvre Brozek J, Roudot A, Staels B, Hum DW, Megnien SJ, Hosmane S,
E. A randomized, placebo-controlled trial of cenicriviroc for treat- Dam N, Chaumat P, Hanf R, Anstee QM, Sanyal AJ. A blood-based
ment of nonalcoholic steatohepatitis with fibrosis. Hepatology 67: biomarker panel (NIS4) for non-invasive diagnosis of non-alcoholic
1754–1767, 2018. doi:10.1002/hep.29477. steatohepatitis and liver fibrosis: a prospective derivation and global
7. Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van validation study. Lancet Gastroenterol Hepatol 5: 970–985, 2020.
Natta ML, Abdelmalek MF, Chalasani N, Dasarathy S, Diehl AM, doi:10.1016/S2468-1253(20)30252-1.
Hameed B, Kowdley KV, McCullough A, Terrault N, Clark JM, 20. Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alco-
Tonascia J, Brunt EM, Kleiner DE, Doo E; NASH Clinical Research holic fatty liver disease: clinical prediction rules and blood-based
Network. Farnesoid X nuclear receptor ligand obeticholic acid for biomarkers. J Hepatol 68: 305–315, 2018. doi:10.1016/j.jhep.2017.
non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, 11.013.
randomised, placebo-controlled trial. Lancet 385: 956–965, 2015 21. De Rubis G, Krishnan SR, Bebawy M. Liquid biopsies in cancer di-
[Erratum in Lancet 385: 946, 2015]. doi:10.1016/S0140-6736(14) agnosis, monitoring, and prognosis. Trends Pharmacol Sci 40: 172–
61933-4. 186, 2019. doi:10.1016/j.tips.2019.01.006.
8. Ratziu V, Harrison SA, Francque S, Bedossa P, Lehert P, Serfaty L, 22. Liu J, Xiao Y, Wu X, Jiang L, Yang S, Ding Z, Fang Z, Hua H, Kirby
Romero-Gomez M, Boursier J, Abdelmalek M, Caldwell S, Drenth MS, Shou J. A circulating microRNA signature as noninvasive diag-
J, Anstee QM, Hum D, Hanf R, Roudot A, Megnien S, Staels nostic and prognostic biomarkers for nonalcoholic steatohepatitis.
B, Sanyal A; GOLDEN-505 Investigator Study Group. Elafibranor, BMC Genomics 19: 188, 2018. doi:10.1186/s12864-018-4575-3.
an agonist of the peroxisome proliferator-activated receptor-a and 23. Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A, Dulai PS,
-d, induces resolution of nonalcoholic steatohepatitis without fibrosis Caussy C, Bettencourt R, Highlander SK, Jones MB, Sirlin CB,
worsening. Gastroenterology 150: 1147–1159, 2016. doi:10.1053/j. Schnabl B, Brinkac L, Schork N, Chen CH, Brenner DA, Biggs W,
gastro.2016.01.038. Yooseph S, Venter JC, Nelson KE. Gut microbiome-based metage-
9. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, nomic signature for non-invasive detection of advanced fibrosis in
Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, human nonalcoholic fatty liver disease. Cell Metab 25: 1054–1062,
Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck 2017. doi:10.1016/j.cmet.2017.04.001.
PR; NASH CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic 24. Van Keuren-Jensen KR, Malenica I, Courtright AL, Ghaffari LT,
steatohepatitis. N Engl J Med 362: 1675–1685, 2010. doi:10.1056/ Starr AP, Metpally RP, Beecroft TA, Carlson EW, Kiefer JA,
NEJMoa0907929. Pockros PJ, Rakela J. microRNA changes in liver tissue associated
10. Lee Y, Doumouras AG, Yu J, Brar K, Banfield L, Gmora S, Anvari M, with fibrosis progression in patients with hepatitis C. Liver Int 36:
Hong D. Complete resolution of nonalcoholic fatty liver disease after 334–343, 2016. doi:10.1111/liv.12919.
bariatric surgery: a systematic review and meta-analysis. Clin 25. Zeybel M, Hardy T, Robinson SM, Fox C, Anstee QM, Ness T,
Gastroenterol Hepatol 17: 1040–1060, 2019. doi:10.1016/j.cgh. Masson S, Mathers JC, French J, White S, Mann J. Differential DNA
2018.10.017. methylation of genes involved in fibrosis progression in non-alco-
11. Romero-Go  mez M, Zelber-Sagi S, Trenell M. Treatment of NAFLD holic fatty liver disease and alcoholic liver disease. Clin Epigenetics
with diet, physical activity and exercise. J Hepatol 67: 829–846, 7: 25, 2015. doi:10.1186/s13148-015-0056-6.
2017. doi:10.1016/j.jhep.2017.05.016. 26. Ryaboshapkina M, Hammar M. Human hepatic gene expression
12. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres- signature of non-alcoholic fatty liver disease progression, a meta-
Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, Friedman SL, analysis. Sci Rep 7: 12361, 2017. doi:10.1038/s41598-017-10930-w.
Diago M, Romero-Gomez M. Weight loss through lifestyle modifica- 27. Koh W, Pan W, Gawad C, Fan HC, Kerchner GA, Wyss-Coray T,
tion significantly reduces features of nonalcoholic steatohepatitis. Blumenfeld YJ, El-Sayed YY, Quake SR. Noninvasive in vivo moni-
Gastroenterology 149: 367–378, 2015. doi:10.1053/j.gastro.2015. toring of tissue-specific global gene expression in humans. Proc
04.005. Natl Acad Sci USA 111: 7361–7366, 2014 [Erratum in Proc Natl Acad
13. Vilar-Gomez E, Yasells-Garcia A, Martinez-Perez Y, Calzadilla- Sci USA 111: 11223, 2014]. doi:10.1073/pnas.1405528111.
Bertot L, Torres-Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, 28. Munchel S, Rohrback S, Randise-Hinchliff C, Kinnings S,
Villa-Jimenez O, Friedman SL, Diago M, Romero-Gomez M. Deshmukh S, Alla N ,Tan C, Kia A, Greene G, Leety L, Rhoa M,
Development and validation of a noninvasive prediction model for Yeats S, Saul M, Chou J, Bianco K, O'Shea K, Bujold R, Norwitz E,
nonalcoholic steatohepatitis resolution after lifestyle intervention. Wapner E, Saade G, Kaper F. Circulating transcripts in maternal
Hepatology 63: 1875–1887, 2016. doi:10.1002/hep.28484. blood reflect a molecular signature of early-onset preeclampsia. Sci
14. Castera L, Friedrich-Rust M, Loomba R. Noninvasive assessment of Transl Med 12: eaaz0131, 2020. doi:10.1126/scitranslmed.aaz0131.
liver disease in patients with nonalcoholic fatty liver disease. 29. Ngo TTM, Moufarrej MN, Rasmussen MH, Camunas-Soler J, Pan
Gastroenterology 156: 1264–1281, 2019. e1264 doi:10.1053/j.gastro. W, Okamoto J, Neff NF, Liu K, Wong RJ, Downes K, Tibshirani R,
2018.12.036. Shaw GM, Skotte L, Stevenson DK, Biggio JR, Elovitz MA, Melbye
15. Nobili V, Parkes J, Bottazzo G, Marcellini M, Cross R, Newman D, M, Quake SR. Noninvasive blood tests for fetal development predict
Vizzutti F, Pinzani M, Rosenberg WM. Performance of ELF serum gestational age and preterm delivery. Science 360: 1133–1136, 2018.
markers in predicting fibrosis stage in pediatric non-alcoholic fatty doi:10.1126/science.aar3819.
liver disease. Gastroenterology 136: 160–167, 2009. doi:10.1053/j. 30. Ibarra A, Zhuang J, Zhao Y, Salathia NS, Huang V, Acosta AD,
gastro.2008.09.013. Aballi J, Toden S, Karns AP, Purnajo I, Parks JR, Guo L, Mason J,
16. Siddiqui MS, Vuppalanchi R, Van Natta ML, Hallinan E, Kowdley Sigal D, Nova TS, Quake SR, Nerenberg M. Non-invasive character-
KV, Abdelmalek M, Neuschwander-Tetri BA, Loomba R, Dasarathy ization of human bone marrow stimulation and reconstitution by cell-
S, Brandman D, Doo E, Tonascia JA, Kleiner DE, Chalasani N, free messenger RNA sequencing. Nat Commun 11: 400, 2020.
Sanyal AJ; NASH Clinical Research Network. Vibration-controlled doi:10.1038/s41467-019-14253-4.
transient elastography to assess fibrosis and steatosis in patients 31. Simons M, Raposo G. Exosomes–vesicular carriers for intercellular
with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 17: communication. Curr Opin Cell Biol 21: 575–581, 2009. doi:10.1016/j.
156–163.e2, 2019. doi:10.1016/j.cgh.2018.04.043. ceb.2009.03.007.

G448 AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.
STRATIFICATION OF NAFLD BY CELL-FREE MRNA CHARACTERIZATION

32. van den Boorn JG, Dassler J, Coch C, Schlee M, Hartmann G. induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med
Exosomes as nucleic acid nanocarriers. Adv Drug Deliv Rev 65: 10: eaat0344, 2018. doi:10.1126/scitranslmed.aat0344.
331–335, 2013. doi:10.1016/j.addr.2012.06.011. 46. Pepe MS, Feng Z, Huang Y, Longton G, Prentice R, Thompson IM,
33. Chang M-L, Yang S-S. Metabolic signature of hepatic fibrosis: from Zheng Y. Integrating the predictiveness of a marker with its perform-
individual pathways to systems biology. Cells 8: 1423, 2019. ance as a classifier. Am J Epidemiol 167: 362–368, 2008.
doi:10.3390/cells8111423. doi:10.1093/aje/kwm305.
34. Hoang SA, Oseini A, Feaver RE, Cole BK, Asgharpour A, Vincent 47. Younossi ZM, Blissett D, Blissett R, Henry L, Stepanova M,
R, Siddiqui M, Lawson MJ, Day NC, Taylor JM, Wamhoff BR, Younossi Y, Racila A, Hunt S, Beckerman R. The economic and
Mirshahi F, Contos MJ, Idowu M, Sanyal AJ. Gene expression pre- clinical burden of nonalcoholic fatty liver disease in the United
dicts histological severity and reveals distinct molecular profiles of States and Europe. Hepatology 64: 1577–1586, 2016. doi:10.1002/
nonalcoholic fatty liver disease. Sci Rep 9: 12541, 2019. doi:10.1038/ hep.28785.
s41598-019-48746-5. 48. Bravo AA, Sheth SG, Chopra S. Liver biopsy. N Engl J Med 344:
35. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, 495–500, 2001. doi:10.1056/NEJM200102153440706.
Cummings OW, Ferrell LD, Liu YC, Torbenson MS, Unalp-Arida A, 49. Ratziu V, Charlotte F, Heurtier A, Gombert S, Giral P, Bruckert E,
Yeh M, McCullough AJ, Sanyal AJ; Nonalcoholic Steatohepatitis Grimaldi A, Capron F, Poynard T; LIDO Study Group. Sampling variabili-
Clinical Research Network. Design and validation of a histological ty of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology
scoring system for nonalcoholic fatty liver disease. Hepatology 41: 128: 1898–1906, 2005. doi:10.1053/j.gastro.2005.03.084.
1313–1321, 2005. doi:10.1002/hep.20701. 50. Wong VW, Adams LA, de Le dinghen V, Wong GL, Sookoian S.
36. Love MI, Huber W, Anders S. Moderated estimation of fold change Noninvasive biomarkers in NAFLD and NASH - current progress and
and dispersion for RNA-seq data with DESeq2. Genome Biol 15: future promise. Nat Rev Gastroenterol Hepatol 15: 461–478, 2018.
550, 2014. doi:10.1186/s13059-014-0550-8. doi:10.1038/s41575-018-0014-9.
37. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, 51. Bianchi DW, Chiu RWK. Sequencing of circulating cell-free DNA
Tanaseichuk O, Benner C, Chanda SK. Metascape provides a during pregnancy. N Engl J Med 379: 464–473, 2018. doi:10.1056/
biologist-oriented resource for the analysis of systems-level NEJMra1705345.
datasets. Nat Commun 10: 1523, 2019. doi:10.1038/s41467-019- 52. De Vlaminck I, Valantine HA, Snyder TM, Strehl C, Cohen G,
09234-6. Luikart H, Neff NF, Okamoto J, Bernstein D, Weisshaar D, Quake
38. Toden S, Zhuang J, Acosta AD, Karns AP, Salathia NS, Brewer JB, SR, Khush KK. Circulating cell-free DNA enables noninvasive diag-
Wilcock DM, Aballi J, Nerenberg M, Quake SR, Ibarra A. Non-inva- nosis of heart transplant rejection. Sci Transl Med 6: 241ra77, 2014.
sive characterization of Alzheimer’s disease by circulating, cell-free doi:10.1126/scitranslmed.3007803.
messenger RNA next generation sequencing. Sci Adv 6: eabb1654, 53. Fan HC, Blumenfeld YJ, Chitkara U, Hudgins L, Quake SR.
2020. doi:10.1126/sciadv.abb1654. Noninvasive diagnosis of fetal aneuploidy by shotgun sequencing
39. Baker SC, Bauer SR, Beyer RP, Brenton JD, Bromley B, Burrill J; DNA from maternal blood. Proc Natl Acad Sci USA 105: 16266–
External RNA Controls Consortium, et al. The external RNA con- 16271, 2008. doi:10.1073/pnas.0808319105.
trols consortium: a progress report. Nat Methods 2: 731–734, 2005. 54. Fan HC, Gu W, Wang J, Blumenfeld YJ, El-Sayed YY, Quake SR.
doi:10.1038/nmeth1005-731. Non-invasive prenatal measurement of the fetal genome. Nature
40. GTEx Consortium. The genotype-tissue expression (GTEx) project. 487: 320–324, 2012 [Erratum in Nature 489: 326, 2012]. doi:10.1038/
Nat Genet 45: 580–585, 2013. doi:10.1038/ng.2653. nature11251.
41. Moylan CA, Pang H, Dellinger A, Suzuki A, Garrett ME, Guy CD, 55. Grskovic M, Hiller DJ, Eubank LA, Sninsky JJ, Christopherson C,
Murphy SK, Ashley-Koch AE, Choi SS, Michelotti GA, Hampton Collins JP, Thompson K, Song M, Wang YS, Ross D, Nelles MJ,
DD, Chen Y, Tillmann HL, Hauser MA, Abdelmalek MF, Diehl AM. Yee JP, Wilber JC, Crespo-Leiro MG, Scott SL, Woodward RN.
Hepatic gene expression profiles differentiate presymptomatic Validation of a clinical-grade assay to measure donor-derived cell-
patients with mild versus severe nonalcoholic fatty liver disease. free DNA in solid organ transplant recipients. J Mol Diagn 18: 890–
Hepatology 59: 471–482, 2014. doi:10.1002/hep.26661. 902, 2016. doi:10.1016/j.jmoldx.2016.07.003.
42. Karsdal MA, Detlefsen S, Daniels SJ, Nielsen MJ, Krag A, 56. Snyder TM, Khush KK, Valantine HA, Quake SR. Universal noninva-
Schuppan D. Is the total amount as important as localization and sive detection of solid organ transplant rejection. Proc Natl Acad Sci
type of collagen in liver fibrosis attributable to steatohepatitis? USA 108: 6229–6234, 2011. doi:10.1073/pnas.1013924108.
Hepatology 71: 346–351, 2020. doi:10.1002/hep.30969. 57. Sumida Y, Yoneda M. Current and future pharmacological therapies
43. Zhong Y, Qin Y, Dang L, Jia L, Zhang Z, Wu H, Cui J, Bian H, Li Z. for NAFLD/NASH. J Gastroenterol 53: 362–376, 2018. doi:10.1007/
Alteration and localization of glycan-binding proteins in human he- s00535-017-1415-1.
patic stellate cells during liver fibrosis. Proteomics 15: 3283–3295, 58. Wong VW, Chitturi S, Wong GL, Yu J, Chan HL, Farrell GC.
2015. doi:10.1002/pmic.201500030. Pathogenesis and novel treatment options for non-alcoholic steato-
44. Reddy JK, Rao MS. Lipid metabolism and liver inflammation. II. Fatty hepatitis. Lancet Gastroenterol Hepatol 1: 56–67, 2016. doi:10.1016/
liver disease and fatty acid oxidation. Am J Physiol Gastrointest Liver S2468-1253(16)30011-5.
Physiol 290: G852–G858, 2006. doi:10.1152/ajpgi.00521.2005. 59. Xiao G, Zhu S, Xiao X, Yan L, Yang J, Wu G. Comparison of labora-
45. Zhu C, Kim K, Wang X, Bartolome A, Salomao M, Dongiovanni P, tory tests, ultrasound, or magnetic resonance elastography to detect
Meroni M, Graham MJ, Yates KP, Diehl AM, Schwabe RF, Tabas I, fibrosis in patients with nonalcoholic fatty liver disease: a meta-anal-
Valenti L, Lavine JE, Pajvani UB. Hepatocyte notch activation ysis. Hepatology 66: 1486–1501, 2017. doi:10.1002/hep.29302.

AJP-Gastrointest Liver Physiol  doi:10.1152/ajpgi.00397.2020  www.ajpgi.org G449


Downloaded from journals.physiology.org/journal/ajpgi (154.080.113.085) on November 18, 2023.

You might also like