HHS Public Access: Dietary Intake and Glutamine-Serine Metabolism Control Pathologic Vascular Stiffness
HHS Public Access: Dietary Intake and Glutamine-Serine Metabolism Control Pathologic Vascular Stiffness
Author manuscript
Cell Metab. Author manuscript; available in PMC 2025 June 04.
Author Manuscript
Sina Tavakoli3,10, Olaf Mercier4,11, Michael G. Risbano2,7, Zhong-Ke Yao12, Guangli Yang12,
Ouathek Ouerfelli12, Jason S. Lewis12, David Montani5,11, Marc Humbert4,11, Matthew L.
Steinhauser8,9, Carolyn Anderson13, William M. Oldham14, Frédéric Perros4,15, Thomas
Bertero1,§,#,$, Stephen Y. Chan2,3,§,#
(1)Université Côte d’Azur, CNRS, INSERM, IPMC, IHU-RespirERA Valbonne, France;
(2)Centerfor Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood
Vascular Medicine Institute,
(3)Division
of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and
UPMC, Pittsburgh, PA, USA;
(4)UniversitéParis–Saclay, AP-HP, INSERM UMR_S 999, Service de Pneumologie et Soins
Intensifs Respiratoires, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
Author Manuscript
(5)Pôle
Thoracique, Vasculaire et Transplantations, Hôpital Marie Lannelongue, Le Plessis-
Robinson, France;
#
Corresponding Authors: Thomas Bertero, Ph.D., Université Côte d’Azur, Institute of Molecular and Cellular Pharmacology,
CNRS UMR 7275 – INSERM U1323, 660, route des Lucioles, Sophia Antipolis, 06560 Valbonne, France, thomas.bertero@univ-
cotedazur.fr, Stephen Y Chan, M.D., Ph.D., Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and
Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and
UPMC, 200 Lothrop Street BST E1240, Pittsburgh, PA USA 15261, chansy@pitt.edu.
*denotes equal contribution by these authors.
§denotes equal contribution by these authors.
$Lead contact
Author contributions: S.Y.C. and T.B. conceived and designed the experiments. S.Y.C., W.M.O., F.P. and T.B., provided
experimental infrastructure. N.R., Y.T., C.C., S.T. C.S., A.A.M., Y-Y.T., J.Z., W.M.O., and T.B. performed the in vitro experiments.
F.B., S.C. and S.A. performed confocal imaging experiments. Y.T., Y-Y.T., J.Z.., J.G., K.F.A., F.P., and S.Y.C. performed the in vivo
Author Manuscript
rat and mouse experiments. L.E., Y.T., Y-Y.T., S.Y.C., and M.L.S. performed the MIMS study. G.Y., Z.-K.Y, and O.O. synthesized the
18F-Gln precursor and FGln standard. Y.T., M.T., J.D.L., S.T., S.Y.C., and C.A. performed the PET imaging studies. Y.A.A., M.G.R.,
and Y.Z. provided human plasma samples and clinical phenotyping data. O.M., D.M., F.P., and M.H., provided human PAAF and
clinical phenotyping data. W.M.O. performed and analyzed the metabolomic experiments. S.Y.C. and T.B. wrote the manuscript. All
authors participated in interpreting the results and revising the manuscript.
Declaration of Interests: S.Y.C. has served as a consultant for Merck, Janssen, and United Therapeutics. S.Y.C. is a director, officer,
and shareholder in Synhale Therapeutics. S.Y.C. has held research grants from WoodNext, Bayer, and United Therapeutics. S.Y.C. and
T.B. have filed patent applications regarding the targeting of metabolism in pulmonary hypertension. G.Y. Z.-K.Y and O.O. are listed
as inventors in patents, not related to this work, that are filed by MSKCC. O.O. receives royalties from MSKCC, Johnson & Johnson,
Jazz, and Y-mAbs and owns shares of Angiogenex for which he is an unpaid member of the SAB, all of which are not related to this
work. The other authors declare no competing interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review
of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered
which could affect the content, and all legal disclaimers that apply to the journal pertain.
Rachedi et al. Page 2
(6)Hillman Cancer Center, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA,
Author Manuscript
USA;
(7)Divisionof Pulmonary and Critical Care Medicine, Department of Medicine, University of
Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA;
(8)Centerfor NanoImaging, Division of Genetics, Brigham and Women’s Hospital and Harvard
Medical School, Boston, MA
(9)AgingInstitute, Division of Cardiology, Department of Medicine, University of Pittsburgh School
of Medicine and UPMC, Pittsburgh, PA, USA;
(10)Department of Radiology, University of Pittsburgh School of Medicine and UPMC, Pittsburgh,
PA, USA;
(11)Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs
Author Manuscript
Summary
Author Manuscript
Perivascular collagen deposition by activated fibroblasts promotes vascular stiffening and drives
cardiovascular diseases such as pulmonary hypertension (PH). Whether and how vascular
fibroblasts rewire their metabolism to sustain collagen biosynthesis remain unknown. Here, we
found that inflammation, hypoxia, and mechanical stress converge on activating the transcriptional
coactivators YAP and TAZ (WWTR1) in pulmonary arterial adventitial fibroblasts (PAAF).
Consequently, YAP and TAZ drive glutamine and serine catabolism to sustain proline and glycine
anabolism and promote collagen biosynthesis. Pharmacologic or dietary intervention on proline
and glycine anabolic demand decreases vascular stiffening and improves cardiovascular function
in PH rodent models. By identifying the limiting metabolic pathways for vascular collagen
biosynthesis, our findings provide guidance for incorporating metabolic and dietary interventions
for treating cardiopulmonary vascular disease.
Author Manuscript
Graphical Abstract
eTOC blurb
Rachedi et al. identify the precise metabolic requirements for pathogenic vascular fibroblast
activation, using pulmonary arterial hypertension as an example. Inflammation, hypoxia, and
Author Manuscript
mechanical stress converge on YAP and TAZ activation. YAP and TAZ control glutamine and
serine catabolism to generate proline and glycine for collagen biosynthesis. Pharmacologic or
dietary intervention on those metabolic pathways decreases collagen production and vascular
stiffening and improves pulmonary vascular function.
Keywords
Cardiovascular Disease; Metabolism; Nutrition; Vascular Fibroblast; Fibrosis; Collagen
Metabolism
INTRODUCTION
Cardiac and vascular diseases (CVDs) constitute the leading cause of death worldwide1.
Author Manuscript
Vascular remodeling and stiffening are hallmarks of CVDs and primarily contribute to
CVD severity2. Dysregulated balance between collagen deposition and degradation has
been reported to promote vascular stiffening. Recently, we and others have shown that
activated vascular fibroblasts promote collagen biosynthesis in CVDs3–5. For instance, in
pulmonary hypertension (PH), a deadly enigmatic disease of the pulmonary vasculature, as
well as in a particularly deadly subtype pulmonary arterial hypertension (PAH), hormonal,
inflammatory, and environmental stresses such as hypoxia/ischemia, or mechanical cues
Under the control of adventitial fibroblasts are the hundreds of ECM proteins and fibrillar
collagens that have been shown to provide the scaffold and give mechanical properties
to the vascular wall structure6–8. Dysregulation of the mechanical properties of vascular
cells microenvironment is increasingly recognized as a driver of aberrant vascular cell
reprogramming. As such changes in vascular collagen metabolism is capable of directly
Author Manuscript
affecting resident vascular cell wall behaviors in a manner that influences overall vascular
tone and wall structure. The amino acid composition of collagen is atypical for proteins.
The most common motifs in the amino acid sequence of collagen are glycine-proline-X
and glycine-X-hydroxyproline, where X is any amino acid other than glycine, proline or
hydroxyproline. As such, around 33% of collagen amino acids are glycine, while proline
and hydroxyproline account for 20% of collagen amino acids9. Consequently, the massive
collagen secretion by activated fibroblasts observed during PH progression must require
these cells to adapt their metabolism to match their energetic needs.
and wet bench research have connected obesity and dietary consumption with increased
CVD risk, while a low carbohydrate, low-fat diet promotes reduced CVD incidence11.
Emerging evidence has emphasized the importance of timed and intermittent dietary
ingestion in influencing global metabolic health in CVD12,13. Yet, the specific dietary
components that control vascular stiffening and downstream cardiopulmonary vascular
phenotypes in health and disease are poorly defined. This is particularly true in pulmonary
vascular diseases, where the impact of diet on risk and severity of PH has not been studied in
depth at a molecular level14.
Here, using metabolomic discovery platforms across primary diseased vascular cells, in vivo
rodent models of PH, and lung tissue of patient with PH, we sought to define the precise
metabolic requirements of pulmonary adventitial fibroblast (PAAF) during pathogenic
Author Manuscript
activation. In doing so, we developed both targeted pharmacologic and dietary interventions
on those metabolic circuits that reduced collagen production and vascular stiffening, thus
improving vascular function in PH in vivo.
RESULTS
Author Manuscript
PAAF activation rewires glutamine and serine metabolism to sustain proline and glycine
biosynthesis.
To determine whether activation of PAAFs rewires cellular metabolism to sustain ECM
production, we performed a series of metabolomic studies on PAAF culture on soft or
stiff matrices and exposed to PH triggers. Here, a soft matrix was defined by a Young’s
(elastic) modulus of 1 kPa, stiff matrix was defined as 12 kPa consistent with our prior
studies of the stiffness of non-diseased and diseased pulmonary arterioles in rodents3,15.
Multiple PH triggers, including exposure to interleukin-6 (IL-6) and transforming growth
factor beta (TGF-β), matrix stiffening (12kPa), and hypoxia, activated PAAFs, as reflected
by increased contractility (increased myosin light chain 2 phosphorylation, P-MLC2; Figure
S1A–C). Steady-state metabolic cell profiles of activated PAAFs (Figure S1D–F) or PAAFs
Author Manuscript
from PH donors (Table S1; Figure 1A and S1G) demonstrated that activation promoted
substantial global alterations of cell metabolism. Consistent with the notion that collagen
production by fibroblasts requires proline and glycine anabolism, pathway analysis revealed
that fibroblast activation significantly altered the steady-state levels of metabolites highly
enriched in pathways related to proline and glycine metabolism (Figures 1B and S1E).
Consistent with these results, the expression levels of several genes involved in proline and
serine biosynthesis were upregulated in activated PAAFs (Figure S1H–I). To determine any
preferential carbon sources for proline and glycine biosynthesis, we sought for metabolites
that are increasingly taken up by activated fibroblasts or diseased lungs. Steady-state
metabolomic analysis of conditioned media from either PH-PAAFs (Figure 1C) or activated-
PAAFs (Figure S1J) revealed a reduction of glutamine, serine, glucose (and, to lesser
extents, proline and glutamate), suggesting that these cells exhibit increased uptake of these
amino acids. Correspondingly, metabolic analysis of the trans-pulmonary plasma gradient
Author Manuscript
To discriminate between carbon sources (Figure 2A) that are taken up by cells from the
media and to determine if at least a portion of the metabolic rewiring observed in activated
PAAFs provides carbons for proline and glycine biosynthesis, we employed stable isotope
tracing studies, using [U-13C]-glucose, [U-13C]-glutamine, [U-13C]-glutamate, or [U-13C]-
serine (Figures 2B–E and S2). Cells were incubated with labeled metabolite for 4 hours.
These tracing experiments demonstrated that glucose, glutamine, and serine catabolism
were increased upon PAAF activation. Moreover, both glutamine-derived proline (M+5) and
Author Manuscript
serine-derived glycine (M+3) levels increased in response to PAAF activation (Figures S2B
and S2D). In contrast, glucose and glutamate did not significantly account for proline and
glycine biosynthesis (Figures S2A and S2C). To derive further insight into the metabolic
rewiring of activated fibroblasts in vivo, we performed complementary experiments
quantifying glutamine and serine uptake. 18F-labeled fluoroglutamine (18F-FGln)16 coupled
with positron emission tomography (PET) scanning correspondingly revealed increased
glutamine absorption by diseased lungs and lung vasculature in monocrotaline-exposed rats
uptake at tissue-scale resolution with glutamine and serine imaging at cellular resolution,
we leveraged a multi-isotope imaging mass spectrometry (MIMS), merging in vivo stable
isotope tracer methodology with nanoscale secondary ion mass spectrometry (Figure 2I–M).
Consistent with the in vitro tracing studies and 18F-FGln PET imaging, significantly elevated
glutamine and serine uptake was observed in the adventitia of diseased pulmonary arterioles
in monocrotaline-exposed PH rats. Together, our results indicate that activated-PAAFs
increase their glutamine and serine uptake, with these amino acids serving as substrates
for production of proline and glycine and consequent collagen biosynthesis and vascular
stiffening in PH.
To investigate whether glutamine and serine catabolism are required for vascular collagen
biosynthesis, we tested whether inhibition of glutaminase (GLS1) (the enzyme that converts
glutamine to glutamate, the first step of glutamine conversion to proline) and/or the serine
hydroxymethyltransferase (SHMT1) (the enzyme that converts the serine to glycine and
is the key isoform as compared to SHMT2 that is responsive to PH triggers, Figure
S1) controls collagen production by activated PAAFs. GLS1 inhibition, achieved via
siRNA (Figure S3A–C) or the small molecule CB-839 (Figure S3D–E), reduced proline
biosynthesis, while SHMT1 inhibition achieved via siRNA or the small molecule SHIN1
blunted glycine biosynthesis. Inhibition of both GLS1 and SHMT1 drastically rewired the
metabolism of activated PAAFs and diseased PAAFs and decreased both proline and glycine
biosynthesis (Figure S3A–E). Consistent with our hypothesis, siRNA knockdown of GLS1
or SHMT1, and to a greater extent siRNA knockdown of GLS1 and SHMT1, decreased
Author Manuscript
collagen production by PAAFs activated by PH triggers such as IL-6 (Figure 3A), stiffness
(Figure S3F) or hypoxia (Figure S3G). Supplementation with permeant α-ketoglutarate
rescues collagen production (Figure S3H). Similar results were obtained in PH-PAAFs
treated with CB-839 and SHIN1 (Figure 3B and S3I).
decreased fibrillar collagen deposition and assembly, thereby decreasing vascular cells
proliferation (Figure 3D–E). Similar results were observed in PH-PAAFs treated with GLS1
and SHMT1 pharmacologic inhibitors (Figure 3F–G). Next, to determine whether the PAEC
and PASMC proliferation observed rely on the mechanical qualities of the collagen-rich
environment or the molecular content we performed a series of complementary experiments.
First, we analyzed the proliferation of vascular cells cultivated on soft or stiff matrices
and supplemented with or without proline and glycine. While matrix stiffening increased
the proliferation of both PAEC and PASMC, proline/glycine supplementation did not
Author Manuscript
affect cell proliferation (Figure S3J). Second, we investigated whether proline and glycine
supplementation can increase the proliferation rate of PAEC or PASMC cultivated on a
matrix from non-activated PAAFs (control-PAAFs). Consistent with our results showing
that proline and glycine did not increase the proliferation of cells cultivated on soft matrix,
supplementation with proline/glycine did not increase the proliferation of vascular cells
(Figure 3I). Together, these results offer evidence against a predominant role for the ECM
molecular content of these specific amino acids in controlling vascular cell proliferation. To
demonstrate the role of matrix stiffness in vascular cell proliferation, we cultivated vascular
cells on matrix produced by IL6-activated fibroblast and treated with or without BAPN, an
inhibitor of lysyl-oxidases (LOXs), the enzymes that crosslink the collagen. Consistent with
previous reports3,15,17, BAPN treatment reduced the matrix stiffness as measured by AFM,
(Figure 3H) and decreased the proliferation of vascular cells plated on top of the matrices
Author Manuscript
(Figure 3I). Together, our results define glutamine and serine catabolism as crucial metabolic
processes, essential for fibroblast-driven vascular stiffening and remodeling.
YAP and TAZ are overarching regulators of vascular remodeling by diseased PAAFs.
To delineate the molecular mechanism controlling vascular remodeling by PAAFs during
PH progression, we sought to identify the overarching molecular regulator(s) of glutamine
and serine catabolism in activated fibroblasts. Recently, we and others have reported the
ability of the co-transcriptional factors YAP and TAZ to control both cell activities and
cell metabolism and thus adjust the metabolic needs of cells with their activities15,18,19.
Furthermore, several studies have reported the involvement of YAP and TAZ in CVD20,21
and fibroblast activation3,22. Therefore, to test whether YAP and TAZ globally control
fibroblast metabolism and especially glutamine/proline and serine/glycine metabolism, we
Author Manuscript
analyzed the reported binding sites of TEADs (the transcriptional factors to which YAP
and TAZ bind to mediate their transcriptional effect23,24) as determined by chromatin
immunoprecipitation and DNA sequencing25,26 (ChIP-seq). Pathway enrichment analysis
revealed that, beyond cell proliferation and DNA/RNA metabolism, TEADs control a
large set of genes involved in cell metabolism and included genes involved in glutamine/
proline and serine/glycine metabolism (Figure 4A). To gain further insights on the role of
YAP/TAZ in the coordination of collagen production and the metabolic rewiring required
for collagen biosynthesis, we searched for TEAD binding sites in promoters of genes
involved in the metabolism-dependent circuit of collagen biosynthesis (Figure 4B–C).
TEAD ChIP seq analysis revealed that TEADs bind upstream to many genes from the
glutamine-to-proline and serine-to-glycine metabolic pathways (Figure 4C). To confirm
these results, we performed YAP ChIP-qPCR experiments in PH-PAAFs (Figure 4D). We
Author Manuscript
demonstrated that YAP binds the promoter region of a large set of genes involved in
glutamine and serine rewiring as well as genes involved in collagen metabolism (Figure 4D).
Moreover, manipulating YAP and TAZ expression (Figure S4A) modulated these genes at
the transcriptional level (Figure S4B) and at the protein level for GLS1 and SHMT1 (Figure
S4A). Together, these results demonstrate that YAP and TAZ coordinate the transcription
of a large pool of genes involved in glutamine to proline and serine to glycine metabolism,
as well as collagen metabolism, indicating that YAP and TAZ may orchestrate collagen
biosynthesis via control of amino acid metabolism.
To investigate more precisely whether YAP and TAZ metabolically control collagen
Author Manuscript
biosynthesis, we analyzed the steady-state metabolic profile of YAP and TAZ knockout
cells (Figures 4E–F and S5A–B). Modulating YAP and TAZ expression in activated PAAFs
promoted a substantial global alteration of cell metabolism. Beyond pathways related to
glucose metabolism, the analysis revealed that siRNA knockdown of YAP and TAZ in
activated-PAAFs modulated the steady-state levels of metabolites highly enriched in proline
and glycine metabolism pathways (Figures 4F and S4B). Consistent with the role of YAP
and TAZ in the control of the glutamine to proline and glycine to serine metabolism,
siRNA knockdown of YAP and TAZ in activated-PAAFs decreased the glutamine-derived
proline (M+5) and the serine-derived glycine (M+3) levels (Figures 4G–H and S5C–F) and
decreased collagen production (Figures 4I and S4C–D). Furthermore, we demonstrated that
manipulating YAP and TAZ expression in activated fibroblasts decreased fibrillar collagen
deposition and assembly in the ECM (Figures 4J–L and S4E), thereby decreasing the
Author Manuscript
proliferation of naive vascular cells (Figures 4J–L and S4E). Together, these results showed
that YAP and TAZ directly control the rewiring of glutamine and serine into proline and
glycine metabolism to sustain collagen biosynthesis.
Next, we investigated whether YAP and TAZ metabolically control collagen biosynthesis
in vascular fibroblast in vivo to build a diseased vascular niche promoting PH. Specific
conditional knockdown of YAP and TAZ in fibroblasts of pulmonary-specific IL-6
transgenic mice (Figure 5A) – an inflammatory mice model of PH27—decreased GLS1
(Figure 5B) and SHMT1 (Figure 5C) expression in the pulmonary arteries, decreased the
pulmonary vascular collagen content (total and fibrillar; Figure 5D–F), thereby decreasing
pulmonary arterial stiffness as measured by atomic force microscopy (AFM; Figure 5G).
Given the alterations occurring in the pulmonary vascular ECM, the deletion of YAP and
TAZ in fibroblasts decreased vascular mural cell proliferation (Figure 5H) and improved
Author Manuscript
downstream histologic (Figure 5I), hemodynamic (Figure 5J), and right ventricular indices
(Figure 5K) of PH. Similar results were observed in a second rodent model of PH where
PH is induced by chronic exposure to hypoxia (Figure S6). Together, our results demonstrate
that YAP and TAZ coordinate the metabolic needs of activated vascular fibroblasts with their
collagen remodeling activities to build a diseased vascular niche that favors vascular mural
cell proliferation.
Targeting proline and glycine anabolism decreases collagen production and vascular
stiffening, thus improving PH in vivo.
We next investigated whether targeting glutamine and serine catabolism is sufficient to
decrease vascular stiffening and improve pulmonary vascular function in a pre-clinical
model of PH. Monocrotaline-exposed rats were treated with either CB-839 or SHIN1 or the
Author Manuscript
combination of inhibitors (Figure 6A). Pharmacologic inhibition of both GLS1 and SHMT1
decreased the pulmonary vascular collagen content (Figure 6B–C), decreased the pulmonary
arterial stiffness (Figure 6D), thus decreasing vascular cell proliferation (Figure 6E). As a
result, GLS1 and SHMT1 inhibition significantly decreased pulmonary arteriolar remodeling
and muscularization (Figure 6F), right ventricular systolic pressure (Figure 6G), and right
ventricular remodeling (Figure 6H). In comparison, while CB-839 alone was effective in
reducing vascular collagen content, vascular stiffness, proliferation as well as histologic and
hemodynamic parameters of PH, SHIN1 alone was only modestly active alone across these
Author Manuscript
parameters. Notably, SHIN1 decreased the total amount of fibrillar collagen in the lungs but
did not modulate the soluble/insoluble ratio (Figure 6B). Together, these drugs demonstrated
a more robust improvement than either drug alone, particularly across collagen quality,
stiffness, proliferation, and muscularization.
Based on these results demonstrating the glutamine and serine metabolic requirement for
collagen biosynthesis in vivo during PH development, we investigated whether decreasing
glutamine and serine bio-availability via dietary intervention (Figure 7A) could reduce
collagen biosynthesis and thus improve cardiopulmonary function. We first studied whether
decreasing glutamine and serine ingestion chronically reduced glutamine and serine
bioavailability (Figure 7B). Indeed, we found rapid and sustained decreases in circulating
levels of glutamine and serine in the plasma of rats fed with glutamine- and serine-free
diets, while displaying minimal or inconsistent impact on glucose (Figure 7B), Second, to
Author Manuscript
assess the effect of dietary serine and glycine (SG) restriction in a pre-clinical PH model,
we transferred 12-week old rats from a normal chow diet to experimental diets (synthetic
diet, Ctrl; or Gln−/ser− synthetic diet) 18 days before monocrotaline injection. Three weeks
later, we analyzed for alterations of amino acid metabolism, vascular stiffening, and PH.
Transferring rats from normal chow diet to an experimental glutamine- and serine-free diet
significantly reduced plasma levels of glutamine and serine (Figure 7C), pulmonary vascular
collagen content (Figure 7D–E), pulmonary arterial stiffness (Figure 7F), and vascular cell
proliferation (Figure 7G–H). Consequently, such an amino-acid restricted diet significantly
decreased pulmonary arteriolar remodeling and muscularization (Figure 7I), right ventricular
systolic pressure (Figure 7J), and right ventricular remodeling (Figure 7K). Together, our
results, offer foundational support for dietary intervention targeting glutamine and serine to
improve pulmonary vascular stiffening, remodeling, and downstream PH.
Author Manuscript
DISCUSSION
Our findings delineate a YAP/TAZ-dependent metabolic circuit controlling collagen
biosynthesis by activated vascular fibroblasts. Specifically, our results establish glutamine
and serine catabolism as crucial steps for collagen biosynthesis. These results carry broad
implications for our fundamental understanding of collagen metabolism - a dysregulated
process in several diseases such as cancer and across CVDs such as PH. Furthermore, our
study reveals a much broader and more complex regulation of the extracellular vascular
niche that could be metabolically targeted. As such, our findings provide scientific guidance
for incorporating pharmacologic and dietary interventions as amino acid-based therapies in
these deadly diseases.
Author Manuscript
Interplay among the transcriptional coactivators YAP and TAZ with cell metabolism has
recently gained traction19,28. YAP and TAZ are activated by various stimuli inherent to the
Hippo signaling pathway, glucose metabolism, as well as mechanical signals. YAP/TAZ
control cell growth, stem cell maintenance, and tissue homeostasis23. Given the energy
requirement of cell proliferation and survival, YAP and TAZ are inhibited when the energy
level is low29–31. Conversely, YAP and TAZ control several metabolic pathways, including
glutaminolysis and anaplerosis, to sustain the metabolic needs of cellular proliferation15,18.
Moreover, YAP and TAZ are activated during tissue repair processes and sustain fibroblast
activation in fibrosis21,22. Consistent with the role of YAP/TAZ and the Hippo signaling
Author Manuscript
pathway in PH3,32, our findings highlight a much more complex regulation of metabolism
by YAP and TAZ in tissue fibrosis – whereby both glutamine and serine catabolism
conspire to generate proline and glycine that ultimately are crucial for de novo synthesis
of collagen. It remains to be seen if glutamine and serine pathways can also act in
a reciprocal feedback loop to further activate YAP/TAZ and reinforce this metabolic
reprogramming in activated PAAFs. Recent data have shown that extracellular uptake rather
than synthesis of proline may be a primary mechanism by which pulmonary vascular
endothelium can generate collagen33. Our data indicate that activated PAAFs – cell types
that carry the bulk of responsibility for collagen deposition – rely more prominently on
proline biosynthesis for new collagen synthesis and vascular stiffening. Given the increasing
appreciation of molecular communication and interdependence between the endothelium
and adventitium34,35, an intriguing mechanism could include a process by which proline
Author Manuscript
Beyond PH, these findings may carry important implications for diseases where ECM
deposition and/or remodeling by tissue-resident fibroblasts drive pathogenesis, such as
peripheral vascular diseases, heart failure with preserved ejection fraction (HFpEF), cancer,
Author Manuscript
and tissue fibrosis (i.e., lung, liver, kidney), in general. Future work is warranted to
define whether non-vascular and vascular fibroblasts in other anatomic compartments
also rely predominantly on glutamine and serine catabolism for collagen synthesis. In
particular, intracellular glutamine content may serve as a central rheostat of diverse cellular
function – one that is even more versatile than currently characterized. For example, recent
findings have emerged regarding the mechano-induced glutamylation and stabilization of
microtubules that can drive cancer cell migration36. Since glutamine levels define a limiting
metabolic requirement for collagen synthesis and fibrosis, an intriguing hypothesis now
may emerge whereby shunting of glutamine to microtubule glutamylation vs. to collagen
deposition may either facilitate metastatic cancer invasion or more dense tissue fibrosis. In
vascular diseases beyond PH, the balance of such fundamental processes may otherwise
define the extent of angiogenesis vs. scar or infarct size across a variety of vascularized
Author Manuscript
Beyond glutamine metabolism, our study emphasized the YAP/TAZ-dependent control over
serine/glycine metabolism. Although often associated with glucose catabolism, here we
demonstrate that glycine biosynthesis relies on exogenous serine catabolism. Whereas non-
activated cells exhibit lower demands for serine and glycine, hyperproliferative cancer cells
demonstrate hyperactive intracellular serine and glycine synthesis and are addicted to de
novo production37,38. Our findings demonstrate that similar metabolic processes are at play
in diseased vasculature, furthering the notion of parallel features of cancer and peripheral
and pulmonary vascular disease39,40. Namely, our findings implicate SHMT1 as a crucial
Author Manuscript
In identifying the crucial metabolic pathways required for collagen biosynthesis in fibrotic
disease, our work offers the potential for non-invasive imaging tools for diagnosis and
prognosis in PH. Despite its morbid prognosis, PH, and particularly PAH, is often
misdiagnosed or ignored, with an average time of >40 months between onset of symptoms
to diagnosis and progression of PAH severity41. A crucial need exists to develop non-
invasive diagnostic imaging tools, particularly that can detect early disease stages and reflect
molecular alterations that drive disease in the lung, vasculature, and right ventricle (RV)42.
While our use of MIMS offered research applicability, this technique is too specialized,
time-consuming, and low-throughput to offer a viable diagnostic platform for widespread
clinical use. In contrast, based on our advanced insights into glutamine metabolism in
Author Manuscript
PH, positron emission tomography (PET) may offer a clinically available platform for
imaging changes in the metabolic state of the vasculature and right ventricle in PH. PET
has been applied in PAH to image 18F-fluorodeoxyglucose (18FDG) uptake as a reflection
of increased glycolysis42 particularly in the RV43. However, this specific PET tracer in
PAH is limited by inconsistent results42 and inability to detect early stages of disease.
Beyond 18FDG, highly glutaminolytic tissue are avid for glutamine, as shown by its uptake
in cancers44, and this process is quantifiable by PET. 18F-(2S,4R)-4-fluoroglutamine (18F-
FGln) is a single stereoisomer analog of glutamine with promising results for imaging of
tumors45 such as gliomas16 and breast cancer44. When coupled with our published findings
that glutamine levels decrease in the pulmonary circulation in human patients with PAH15,
our preclinical data with this tracer strengthen the notion that 18F-FGln and glutamine
uptake could be a feasible modality to track in patients with PAH. Given the dearth of
Author Manuscript
effective imaging tests for PAH and PH in general, a first-in-human trial of 18F-FGln PET in
PH could offer a translational avenue to pursue glutamine imaging to define disease severity
and risk, including at early disease time points. 18F-FGln PET could offer a clinical trial
outcome surrogate, reflecting the molecular pathogenesis of PH. It could also aid in clinical
trial recruitment of “super-responder” patients with PH, thus decreasing patient numbers for
recruitment and potentiating an era of precision medicine for PH.
Coupled with furthering such non-invasive platforms of identifying patients with PH avid
for glutamine uptake, our findings also offer a roadmap toward more effective and precise
pharmacologic therapies for PH. Previously we have shown that glutaminolysis supports
PAEC and PASMC proliferation, migration, and contractility via anaplerosis. By now
identifying glutamine metabolism as key step for collagen biosynthesis in PAAF, our
Author Manuscript
findings highlight the pleiotropic activity of glutamine across the pulmonary vasculature.
It remains unclear if serine metabolism carries as many roles, perhaps serving as an
explanation as to why targeting GLS1 appears consistently more effective than targeting
serine metabolism alone. Yet, the interconnected nature of these metabolic pathways opens
up the possibility of combinatorial therapies. Namely, while isolated inhibition of GLS1
can improve PAH15, the synergism of inhibiting both YAP1 and GLS1 in PH has been
described46. We now further define the therapeutic benefit of simultaneous delivery of GLS1
and SHMT1 inhibitors in the diseased pulmonary vasculature. These findings set the stage
for the clinical development of combination of such synergistic agents for more robust
Author Manuscript
Finally, by defining the limiting carbon sources for collagen biosynthesis, our study reveals
a unique opportunity to introduce dietary modulation in the management of PH and CVDs,
in general. It is well known that dietary changes play a dominant role49,50 in a number of
CVDs. Beyond CVDs, dietary intervention has been proposed to ameliorate tissue fibrosis,
such as in fatty liver diseases51. Moreover, altering the timing of daily food ingestion in
the form of intermittent fasting has increasingly been identified as an effective modulator
of cellular and physiologic metabolic parameters, with robust implications on aging and a
Author Manuscript
variety of chronic diseases52. More recently, intermittent fasting has been proposed as a
potential intervention for right ventricular dysfunction in PAH14. However, a diet aimed at
modulating specific ingested amino acid content has not been tested to our knowledge as
a precise therapeutic intervention for fibrosis or cardiopulmonary diseases. Although both
glutamine and serine (as well as proline and glycine) can be synthesized from sources
other than the diet, our preclinical data indicate that targeted dietary restriction of glutamine
and serine are effective in reducing these amino acid levels in lung tissue, suggesting a
decrease of their overall bioavailability. Future studies are warranted to determine whether
dietary sources of these amino acids predominantly and directly contribute to lung vascular
fibrosis in PH or whether dietary alterations may also serve as a primary but indirect trigger
to altered specific amino acid production and/or degradation in lung as PH progresses.
Furthermore, it is possible that genetic variability in the enzymes that transport and
Author Manuscript
metabolize these dietary amino acids in the gut may influence the therapeutic effects of
such dietary restriction. Translating this approach to human patients would also necessitate
greater insight into the complex cooking and processing that could alter the amino acid
composition of any prescribed dietary prescription. Progress toward network medicine53 and
artificial intelligence could help us to systematically define the complex link between the
diet with the genome and disease and thus prepare for feasible dietary interventions in PH
and beyond.
In sum, we identify the precise metabolic and amino acid requirements for vascular
fibroblast activity. This study provides a fundamental insight into the regulation of collagen
metabolism and fibrosis, which is critical for managing PH and holds relevance for other
fibrotic diseases across other CVDs and beyond. Moreover, our study offers the potential for
Author Manuscript
metabolic PET imaging platforms for more effective non-invasive diagnostic and prognostic
testing in PH and vascular stiffening. It also guides developing pharmacologic and dietary
therapies targeting metabolic crosstalk in the diseased vasculature.
Limitations of study:
Limitations to this study should be acknowledged. When interpreting transpulmonary
gradient analyses from rats and humans, we acknowledge that streaming from hepatic veins
may confound collection from either the inferior vena cava (rat) or right atria (human). To
Author Manuscript
demonstrate the role of YAP and TAZ in vascular fibroblasts in vivo, a cell type-enriched
deletion of YAP and TAZ was accomplished using mice expressing the Cre transgene
driven by a tamoxifen-inducible Col1a2 promoter. Although Col1a2 is primarily expressed
in fibroblasts54, smooth muscle cells also express this gene. Thus, we cannot rule out that
at least a portion of the downstream phenotype may be driven by smooth muscle deletion
of YAP and TAZ and that cell type’s contribution to collagen deposition. Our experiments
with dietary restriction also do not account for the kinetics of endogenously produced
and non-diet administered amino acids, which could confound an interpretation of direct
cause-effect in diet restriction with PH. More broadly, our study is not set up to define
the dynamic and likely bidirectional contribution of total metabolite content across the
plasma, lung vasculature, and other tissue or dietary sources. To address this point, future
metabolomic studies of the lung in PAH could be envisioned that are designed similarly to
prior work quantifying fuel use in the failing human heart55.
Author Manuscript
STAR METHODS
RESOURCE AVAILABILITY
Lead contact—Further information and requests for resources and reagents should
be directed to and will be fulfilled by the Lead Contact, Thomas Bertero, PhD
(thomas.bertero@univ-cotedazur.fr).
• All of the data supporting this study are included in the article. All raw data used
to generate the figures throughout the manuscript can be found within the Data
S1 document
• Any additional information required to reanalyze the data reported in this paper
is available from the lead contact upon request.
with 10% fetal bovine serum (Gibco), Glutamine (2mM, Gibco) and Penicillin/Streptomycin
(1%, Gibco). A triple gas incubator set at 1% oxygen, 94% nitrogen, and 5% carbon dioxide
was used for hypoxic conditions. PAAFs from patients with PH (PH-PAAF) were isolated
from explanted lungs at Marie Lannelongue Hospital. To isolate PAAFs, vascular cells
from dissected pulmonary arteries were cultured in DMEM supplemented with 0.5% FCS.
In this condition, PH-PAAFs that outgrew PAEC and PASMC in culture were isolated
by differential trypsinization. These cells were cultured in DMEM supplemented with
expression as well as by their ability to contract a collagen I rich gel in the absence of
serum. PH-PAAFs from at least five different patients were randomly used throughout
the experiments. All cells were grown in collagen-coated plastic (50 μg/mL) at 37°C in
a humidified 5% CO2 atmosphere. Experiments were performed at passages 2–10. All
cells were routinely tested for mycoplasma by PCR and were carefully maintained in a
centralized cell bank. Primary human (Lonza) pulmonary arterial endothelial cells (PAECs)
were grown in EBM-2 basal medium supplemented with EGM-2 MV BulletKit (Lonza).
Primary human (Lonza) pulmonary arterial smooth muscle cells (PASMCs) were grown in
SmGM-2 (Lonza) For the studies dependent on matrix stiffness, collagen-coated hydrogel
pre-plated in culture wells (Matrigen) was generated from a mix of acrylamide and bis-
acrylamide coated with collagen. Cells were cultured, passaged, and harvested while on top
Author Manuscript
Human studies—Patients with PAH from whom PAAFs were isolated were part of the
French Network on Pulmonary Hypertension, a program approved by our institutional
Ethics Committee, and gave written informed consent (Protocol N8CO-08-003, ID RCB:
2008-A00485-50, approved on June 18, 2008). Patients with PAH and controls from whom
plasma was obtained were part of a University of Pittsburgh School of Medicine cohort.
All patients gave written informed consent. The protocol was approved by the institutional
review board at the University of Pittsburgh (IRB: STUDY19050364).
Rodent models of PH
• Inducible cell-specific YAP and TAZ knockout mice. Col1a2-Cre-ER mice were
crossed onto Yap1fl/fl/Tazfl/fl mice. Tamoxifen was diluted at 20 mg/ml in
corn oil (Sigma). Three daily intraperitoneal injection (IP) doses of 30 mg/kg
of tamoxifen were administered to ten-week-old mice (C57Bl6) in order to
knockout YAP and TAZ in cells expressing Col1a2 – a marker enriched in
fibroblasts. Two weeks after injection, exposure to hypoxia commenced.
for 3 weeks. In all cohorts, on day 21 after hypoxia exposure, right heart
catheterization was performed followed by harvesting of lung tissue, paraffin
embedding, or cryopreservation with OCT (Sigma-Aldrich).
rats, aged 10–14 weeks, were divided into two groups: the PH rats, which received an
injection of 60 mg/kg monocrotaline, and the control rats, which received a saline injection.
After a three-week period, 200 μl of blood was collected from the left common carotid artery
and the inferior vena cava into EDTA tubes. The blood samples were then centrifuged, and
the resulting plasma was stored at −80°C for future use.
Inhibition of GLS1 and/or SHMT1 in PH rats.: To induce PH, male Sprague-Dawley rats
(10–14 weeks old) were injected i.p. with 60 mg/kg monocrotaline (Sigma-Aldrich). After 7
days, serial i.p. injections were given daily of CB-839 (10 mg/kg; Selleck Chemicals) and/or
SHIN1 (5mg/kg; Cayman chemical) versus vehicle control. In all cohorts, on day 21 after
monocrotaline injection, right heart catheterization was performed followed by harvesting of
lung tissue, paraffin embedding, or cryopreservation with OCT (Sigma-Aldrich).
Author Manuscript
Diets.: On standard chow, dietary amino acids are derived from whole proteins contained
in the raw ingredients (wheat, wheatfeed, barley, de-hulled extracted toasted soya, maize
and fish meal), with a small amount of purified lysine added as a supplement. Two
sets of experimental diets were used, both based on Purified Amino Acid Diet from
SAFE Nutrition Service: control synthetic diet contained all essential amino acids plus
serine, glutamate, proline, and glycine; Glutamine and serine-free diet was the same as
synthetic diet-control, but without serine, glutamate, proline, and glycine, with the other
amino acid levels increased proportionally to achieve the same total amino acid content.
From weaning, rats received standard chow. Male Sprague-Dawley rats, aged 10–14
weeks, were assigned to three distinct groups: standard chow; synthetic control diet; or
glutamine and serine-free diet. After 18 days, Sprague-Dawley rats were injected i.p. with
60 mg/kg monocrotaline (Sigma-Aldrich). Right heart catheterization was conducted at day
Author Manuscript
Human plasma harvesting: To collect blood from subjects from the main pulmonary
artery, clinically indicated right heart catheterization procedures were performed by standard
protocol via a right internal jugular approach under fluoroscopic guidance. The catheter was
positioned into the main pulmonary artery, as confirmed by fluoroscopy and hemodynamic
waveforms. Blood was drawn from the distal port and collected in vacutainer tubes with K+-
EDTA. Plasma was extracted after centrifugation of blood, followed by storage at −80°C.
METHOD DETAILS
Plasmids, antibodies, and reagents—The YAP1 coding sequence was purchased
Author Manuscript
Immunoblot assays—Forty-eight hours after plaiting, cells were lysed in RIPA buffer
(Pierce) or directly in Laemmli’s buffer. After denaturation, protein lysates were resolved by
SDS-PAGE and transferred onto a PVDF membrane (Millipore). Membranes were blocked
with 2% BSA in TBS tween20 0.1% and incubated in the presence of the primary and
then secondary antibodies. After washing, immunoreactive bands were visualized with ECL
(Millipore) and analyzed on Fusion-FX Imager (Vilber).
minor modifications. Briefly, metabolites were extracted from cultured cells on dry ice
using 80% aqueous methanol precooled at −80°C. Insoluble materials from both cell and
supernatant extractions was removed by centrifugation at 20,000 g for 15 minutes at
4°C. The supernatant was evaporated to dryness by SpeedVac at 42 °C, the pellet was
resuspended in LC-MS water, and metabolites were analyzed by LC-MS.
acetonitrile. The injection volume was 1 μL, the mobile phase flow rate was 100 μL/min, the
column compartment temperature was set at 25 °C, and the autosampler compartment was
set at 4 °C. The mobile phase gradient (%B) was 0 min, 80%; 5 min 80%; 30 min, 20%; 31
min, 80%; 42 min, 80%. The column effluent was introduced to the mass spectrometer with
the following ionization source settings: sheath gas 40, auxillary gas 15, sweep gas 1, spray
voltage +/− 3.0 kV, capillary temperature 275 °C, S-lens RF level 40, probe temperature 350
°C. The mass spectrometer was operated in polarity switching full scan mode from 70–1000
m/z. Resolution was set to 70,000 and the AGC target was 1×106 ions. Data were acquired
and analysed using TraceFinder software (Thermo) with peak identifications based on an
Author Manuscript
in-house library of authentic metabolite standards previously analysed utilizing this method.
For all metabolomic experiments, the quantity of the metabolite fraction analysed was
adjusted to the corresponding cell number calculated upon processing a parallel experiment.
80% ice-cold methanol (−80°C) and transferred to −80°C freezer for 20 minutes. Cell plates
were resuspended on dry ice and the collected lysate/methanol suspension was transferred
to 15 ml conical tubes kept on dry ice. Suspension was centrifuged at full speed (21,000
g) for 5 minutes at 4°C. Supernatants were then transferred to 50 ml conical tubes on dry
ice. The pellets were resuspended with 500 μl 80% methanol (−80°C) and a combination of
vortexing and pipetting up and down. The suspension was centrifuged at full speed (21,000
g) for 5 minutes at 4°C. Supernatant were transferred to 50 ml conical tubes on dry ice.
Pellets obtained after centrifugation were dissolved in urea 8 M (Tris 10 mM pH 8.0) at
60°C, to later measure protein concentration for normalization. After pooling the metabolite
extractions, the samples are completely dried under a nitrogen gas apparatus (N-EVAP) and
submitted for LC-MS analysis.
Author Manuscript
was collected at a resolution of 70,000. The automatic gain control (AGC) target was set
at 1 × 106 and the maximum injection time was 200 ms. The top 5 precursor ions were
fragmented, in a data-dependent manner, using the higher energy collisional dissociation
(HCD) cell set to 30% normalized collision energy in MS2 at a resolution power of 17,500.
The sample volumes of 10 μl were injected. Data acquisition and analysis were performed
by Xcalibur 4.0 software and Tracefinder 2.1 software, respectively (both from Thermo
Fisher Scientific).
space, the volumetric ROI was thresholded to give representations of both airspace and
bronchovasculature. The SUVmean was determined for each tissue compartment. The SUV
values in these regions were each normalized against the SUVmeanin muscle.
lower bound of the scale (blue) is set at natural background. The upper bound is set to
visually demonstrate differential labeling. Scaling changes affect the color pattern; however,
the underlying quantitative isotopic ratio data is unaffected. 15N-glutamine image acquisition
(Figure 2I–J) was performed with ‘mosaic imaging.’ Alternatively, targeted vessel imaging
was pursued in Figure 2K–L. Specifically, vessels were identified based on size, the
coordinates were recorded, and higher resolution images were then acquired by directly
analyzing individual vessels without capturing surrounding lung parenchyma. This was done
to minimize analytical ‘dead space.’
ChIP-qPCR—PH-PAAF were cultivated on plastic for 48h. Cells were dual cross-linked
Author Manuscript
in the low- and high-salt buffers. After crosslinking reversion (65°C for 4 hours) and
Proteinase K treatment, chromatin was purified by phenol-chloroform extraction and
ethanol precipitation. Precipitated DNA was analyzed by qPCR using primers generated
for predicted TEAD binding sites or a non-relevant genomic region (Control).
coordinates. The promoter region is shown in gray and the gene region is shown in black.
Calculated peaks are shown in red. Hypergeometric enrichment p-values were calculated in
Python 3.8 using the scipy package.
KEGG Analysis—KEGG gene sets were downloaded from the Broad Institute’s
Molecular Signatures Database (MSigDB). Hypergeometric p-values for enrichment of gene
sets for predicted TEAD targets (as defined above) were calculated in Python 3.8 using the
scipy package.
(37°C) extraction buffer (PBS/0.5% Triton X100/20 mM NH4OH). Matrices were washed
3 times (PBS). Naïve PAECs or PASMCs were then plated on top of these matrices
supplemented with normal growth medium 0.5% FBS or harvested for assessment of
collagen (Sircol assay) or fibrillar collagen content (Picrosirius Red stain). Twenty-four
hours later, cells were harvested for immunofluorescence experiments.
for 10 min and permeabilized with PBS/Triton 100X 0.1% for 10min. Then cells were
incubated with anti-P-MLC2 (#3671; 1/100; Cell signaling) or anti-Ki67 (ab15580; 1/300;
Abcam) at room temperature for 2 hours. Secondary antibodies coupled with Alexa-594
(Thermo Scientific) or Alexa-488 were used at 1:500. Nuclei were counterstained with
DAPI (Sigma-Aldrich). Pictures were obtained using a ZEISS LSM Exciter confocal
microscope.
a previously published protocol3. Rodent lung was weighed, minced, and incubated in
0.5 M acetic acid at 4°C. After overnight digestion, the acetic acid-soluble and insoluble
fractions were isolated by centrifugation. The soluble fraction was stored at −80°C, while
the insoluble fraction was digested by overnight incubation in 6M hydrochloric acid at 85°C.
Concentrations of soluble and insoluble (gelatinous) collagen fractions were determined
using a Sircol Soluble Collagen Assay Kit (Biocolor) with a colorimetric reaction (measured
at 550 nm) and a collagen reference standard curve.
Picrosirius red stain and quantification—Picrosirius red stain was achieved through
the use of 5μm paraffin sections stained with 0.1% picrosirius red (Direct Red80, Sigma-
Aldrich) and counterstained with Weigert’s hematoxylin to reveal fibrillar collagen. The
sections were then serially imaged using with an analyzer and polarizer oriented parallel
Author Manuscript
and orthogonal to each other. Microscope conditions (lamp brightness, condenser opening,
objective, zoom, exposure time, and gain parameters) were constant throughout the imaging
of all samples. A minimal threshold was set on appropriate control sections for each
experiment in which only the light passing through the orthogonally-oriented polarizers
representing fibrous structures (i.e., excluding residual light from the black background) was
included. The threshold was maintained for all images across all conditions within each
experiment. The area of the transferred regions that was covered by the thresholded light
was calculated and at least 10 sections/vessel per condition were averaged together (Image J
software).
μm thickness) were cut out from their glass slide and the fragment of glass containing the
sample was glued on the bottom of a 50 mm dish (Willco Glass Bottom Dish). Before
measurements the sample was first rinsed and after covered with 4 ml of PBS 1x. The
mechanical properties of the samples were studied using a BioScope Catalyst atomic force
microscope (Bruker) coupled with and optical microscope (Leica DMI6000B) that enables,
by phase contrast, to pinpoint the areas of interest. For each sample, 8–12 vessels per
rodent (< 100 μm diameter) were analyzed using the “Point and Shoot” method, collecting
from 80 to 120 force-distance curves at just as many discrete points. The experiments of
Author Manuscript
microindentation were performed in PBS using a probe with a Borosilicate Glass spherical
tip (5 μm of diameter) and a cantilever with a nominal spring constant of 0.06 N/m
(Novascan). Indentations were carried out using a velocity of 2 μm/s, in relative trigger
mode and by setting the trigger threshold to 2 nN. The apparent Young’s (elastic) modulus
was calculated using the NanoScope Analysis 1.50 software (Bruker), fitting the force
curves to the Hertz spherical indentation model and using a Poisson’s ratio of 0.4. To
avoid large indentation, a minimum and a maximum Force Fit Boundary of 5% and 25%
respectively of the whole force curve was taken into account for the fit.
of data distribution was determined by Shapiro Wilk testing. Means of 2 sample groups
were compared by paired (when appropriate) and unpaired two-tailed Student’s T-test
for normally distributed data, while Mann-Whitney U non-parametric testing was used
for non-normally distributed data. For comparisons among different groups, analysis of
variance tests (ANOVA) followed by Bonferroni’s post-hoc analysis or Kruskal Wallis tests
with Dunn’s post-hoc analysis (where appropriate for non-normally distributed data) were
performed. A P-value less than 0.05 was considered significant.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Author Manuscript
Acknowledgements:
We thank the B. Mari team members for advice and discussions as well as F. Aguila for artwork. The authors
acknowledge the “Microscopie Imagerie Côte d’Azur” (MICA), GIS-IBISA multi-sites platform and particularly
the IPMC, C3M and IRCAN (Molecular and Cellular Imaging facility PICMI) partners. This platform is supported
by the GIS IBiSA, Conseil Départemental 06, Région PACA ARC, Cancerôpole PACA,)”. We thank the Organic
Synthesis Core (Dr George Sukenick and Ms. Rong Wang) and the NMR Analytical Core (MSKCC) for help with
NMR and LC-MS experiments.
Funding:
This work was supported by: the French National Research Agency ANR-18-CE14-0025, ANR-21-CE44-0036 (for
T.B.) and ANR-20-CE14-0006 (for T.B. and F.P.); NIH grants R01 HL124021, HL122596, HL151228 as well as
the AHA grant 18EIA33900027 (S.Y.C.). The Organic Synthesis Core at MSKCC was supported in part by NCI
R50 CA243895-03 and NCI P30 CA008748-55.
Author Manuscript
REFERENCES
1. Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, Barengo NC,
Beaton AZ, Benjamin EJ, Benziger CP, et al. (2020). Global Burden of Cardiovascular Diseases and
Risk Factors, 1990–2019: Update From the GBD 2019 Study. J Am Coll Cardiol 76, 2982–3021.
10.1016/j.jacc.2020.11.010. [PubMed: 33309175]
2. Cecelja M, and Chowienczyk P (2012). Role of arterial stiffness in cardiovascular disease. JRSM
Cardiovasc Dis 1. 10.1258/cvd.2012.012016.
3. Bertero T, Cottrill KA, Lu Y, Haeger CM, Dieffenbach P, Annis S, Hale A, Bhat B, Kaimal
V, Zhang Y-Y, et al. (2015). Matrix Remodeling Promotes Pulmonary Hypertension through
4. Kothapalli D, Liu S-L, Bae YH, Monslow J, Xu T, Hawthorne EA, Byfield FJ, Castagnino P, Rao S,
Rader DJ, et al. (2012). Cardiovascular protection by apoE and apoE-HDL linked to suppression of
ECM gene expression and arterial stiffening. Cell Rep 2, 1259–1271. 10.1016/j.celrep.2012.09.018.
[PubMed: 23103162]
5. Zhang H, Wang D, Li M, Plecitá-Hlavatá L, D’Alessandro A, Tauber J, Riddle S, Kumar S, Flockton
A, McKeon BA, et al. (2017). Metabolic and Proliferative State of Vascular Adventitial Fibroblasts
in Pulmonary Hypertension Is Regulated Through a MicroRNA-124/PTBP1 (Polypyrimidine
Tract Binding Protein 1)/Pyruvate Kinase Muscle Axis. Circulation 136, 2468–2485. 10.1161/
CIRCULATIONAHA.117.028069. [PubMed: 28972001]
6. Mouw JK, Ou G, and Weaver VM (2014). Extracellular matrix assembly: a multiscale
deconstruction. Nat. Rev. Mol. Cell Biol. 15, 771–785. 10.1038/nrm3902. [PubMed: 25370693]
7. Xu J, and Shi G-P (2014). Vascular wall extracellular matrix proteins and vascular diseases.
Biochim Biophys Acta 1842, 2106–2119. 10.1016/j.bbadis.2014.07.008. [PubMed: 25045854]
8. WAGENSEIL JE, and MECHAM RP (2009). Vascular Extracellular Matrix and Arterial Mechanics.
Author Manuscript
14. Prisco SZ, Eklund M, Moutsoglou DM, Prisco AR, Khoruts A, Weir EK, Thenappan T, and Prins
KW (2021). Intermittent Fasting Enhances Right Ventricular Function in Preclinical Pulmonary
Arterial Hypertension. J Am Heart Assoc 10, e022722. 10.1161/JAHA.121.022722. [PubMed:
34747187]
15. Bertero T, Oldham WM, Cottrill KA, Pisano S, Vanderpool RR, Yu Q, Zhao J, Tai Y, Tang Y,
Zhang Y-Y, et al. (2016). Vascular stiffness mechanoactivates YAP/TAZ-dependent glutaminolysis
to drive pulmonary hypertension. J. Clin. Invest 126, 3313–3335. 10.1172/JCI86387. [PubMed:
27548520]
16. Venneti S, Dunphy MP, Zhang H, Pitter KL, Zanzonico P, Campos C, Carlin SD, La
Rocca G, Lyashchenko S, Ploessl K, et al. (2015). Glutamine-based PET imaging facilitates
enhanced metabolic evaluation of gliomas in vivo. Sci Transl Med 7, 274ra17. 10.1126/
scitranslmed.aaa1009.
17. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A,
Weninger W, et al. (2009). Matrix crosslinking forces tumor progression by enhancing integrin
Author Manuscript
20. Zheng A, Chen Q, and Zhang L (2022). The Hippo-YAP pathway in various cardiovascular
diseases: Focusing on the inflammatory response. Front Immunol 13, 971416. doi: 10.3389/
Author Manuscript
26. Partridge EC, Chhetri SB, Prokop JW, Ramaker RC, Jansen CS, Goh S-T, Mackiewicz M,
Newberry KM, Brandsmeier LA, Meadows SK, et al. (2020). Occupancy maps of 208 chromatin-
associated proteins in one human cell type. Nature 583, 720–728. 10.1038/s41586-020-2023-4.
[PubMed: 32728244]
27. Steiner MK, Syrkina OL, Kolliputi N, Mark EJ, Hales CA, and Waxman AB (2009). Interleukin-6
overexpression induces pulmonary hypertension. Circ Res 104, 236–244, 28p following 244.
10.1161/CIRCRESAHA.108.182014. [PubMed: 19074475]
28. Romani P, Valcarcel-Jimenez L, Frezza C, and Dupont S (2020). Crosstalk between
mechanotransduction and metabolism. Nat Rev Mol Cell Biol 1–17. 10.1038/s41580-020-00306-
w. [PubMed: 31676888]
29. Enzo E, Santinon G, Pocaterra A, Aragona M, Bresolin S, Forcato M, Grifoni D, Pession A,
Zanconato F, Guzzo G, et al. (2015). Aerobic glycolysis tunes YAP/TAZ transcriptional activity.
EMBO J 34, 1349–1370. 10.15252/embj.201490379. [PubMed: 25796446]
30. Mo J-S, Meng Z, Kim YC, Park HW, Hansen CG, Kim S, Lim D-S, and Guan K-L (2015). Cellular
energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat. Cell Biol
Author Manuscript
37. Amelio I, Cutruzzolá F, Antonov A, Agostini M, and Melino G (2014). Serine and glycine
metabolism in cancer. Trends Biochem Sci 39, 191–198. 10.1016/j.tibs.2014.02.004. [PubMed:
Author Manuscript
24657017]
38. Geeraerts SL, Heylen E, De Keersmaecker K, and Kampen KR (2021). The ins and outs of
serine and glycine metabolism in cancer. Nat Metab 3, 131–141. 10.1038/s42255-020-00329-9.
[PubMed: 33510397]
39. DiRenzo D, Owens GK, and Leeper NJ (2017). Attack of the Clones. Circ Res 120, 624–626.
10.1161/CIRCRESAHA.116.310091. [PubMed: 28209794]
40. Boucherat O, Vitry G, Trinh I, Paulin R, Provencher S, and Bonnet S (2017). The cancer theory of
pulmonary arterial hypertension. Pulm Circ 7, 285–299. 10.1177/2045893217701438. [PubMed:
28597757]
41. Hoeper MM, Humbert M, Souza R, Idrees M, Kawut SM, Sliwa-Hahnle K, Jing Z-C, and Gibbs
JSR (2016). A global view of pulmonary hypertension. Lancet Respir Med 4, 306–322. 10.1016/
S2213-2600(15)00543-3. [PubMed: 26975810]
42. van de Veerdonk MC, Marcus JT, Bogaard H-J, and Vonk Noordegraaf A (2014). State of the
art: advanced imaging of the right ventricle and pulmonary circulation in humans (2013 Grover
Author Manuscript
[PubMed: 34056915]
47. Acharya AP, Clare-Salzler MJ, and Keselowsky BG (2009). A high-throughput microparticle
microarray platform for dendritic cell-targeting vaccines. Biomaterials 30, 4168–4177. 10.1016/
j.biomaterials.2009.04.032. [PubMed: 19477505]
48. Roscigno R, Vaughn T, Anderson S, Wargin W, Hunt T, and Hill NS (2020). Pharmacokinetics
and tolerability of LIQ861, a novel dry-powder formulation of treprostinil. Pulm Circ 10,
2045894020971509. 10.1177/2045894020971509. [PubMed: 33282202]
49. Schroeder SA (2007). We Can Do Better — Improving the Health of the American People. New
Engl J Med 357, 1221–1228. 10.1056/NEJMsa073350. [PubMed: 17881753]
50. Khera AV, Emdin CA, Drake I, Natarajan P, Bick AG, Cook NR, Chasman DI, Baber U, Mehran
R, Rader DJ, et al. (2016). Genetic Risk, Adherence to a Healthy Lifestyle, and Coronary Disease.
New Engl J Med 375, 2349–2358. 10.1056/NEJMoa1605086. [PubMed: 27959714]
51. Ozlu T, Yilmaz Y, and Gunes FE (2022). The effects of dietary intervention on fibrosis and
biochemical parameters in metabolic-associated fatty liver disease. Minerva Gastroenterol (Torino)
Author Manuscript
55. Murashige D, Jang C, Neinast M, Edwards JJ, Cowan A, Hyman MC, Rabinowitz JD, Frankel DS,
and Arany Z (2020). Comprehensive quantification of fuel use by the failing and nonfailing human
Author Manuscript
Highlights
Author Manuscript
Figure 1: In vitro and in vivo evidence of proline and glycine metabolic rewiring in PH.
Author Manuscript
(A-B) Heatmap (A) and pathway enrichment analysis (B) of significantly (FDR<0.1;
P<0.05) modulated intracellular metabolites in PH-PAAFs, with log2(fold change) range
between −4 to 4. (C) Heatmap of significantly (FDR<0.1; P<0.05) modulated metabolites
in PH-PAAF conditioned media, with log2(fold change) range between −2 to 2. (D)
Schema of blood sampling for measurement of transpulmonary gradient of metabolites
in rat (yellow, site of blood collection). (E) Heat map of significantly (FDR<0.1; P<0.05)
modulated metabolites in the transpulmonary gradient of monocrotaline-exposed rats (N=8
per group), with log2(fold change) range between −1 to 1. (F) Schema of blood sampling for
Author Manuscript
Figure 2: Glutamine and serine catabolism sustain proline and glycine anabolism in activated
Author Manuscript
as compared with PBS control (N=6). Data are expressed as standard uptake value ratios
Author Manuscript
(SUVr). (I-M) After treating monocrotaline-exposed rats vs. controls (N=3–4 per group)
with 15N-labeled glutamine, 15N-labeled serine, or 15N-labeled serine and 2H-labeled
glutamine administered simultaneously, rats were sacrificed, and fixed lung was analyzed
by multi-isotope imaging mass spectrometry (MIMS). Representative images (I, K and L)
and quantification of 15N-glutamine (J), and 15N-serine and 2H-glutamine (M) incorporation
by pulmonary arteriolar mural cells, expressed as percent above background ratio. The
rainbow scale is set from 0% above background ratio in blue to 100% for glutamine or
150% for serine in red (2 to 2.5 fold above background ratio). Scale=10 μm. In all the
panels *P<0.05; **P<0.01; two-tailed Student’s t-test was used; data show mean ± s.e.m. N’
numbers indicate biological replicates.
Author Manuscript
Author Manuscript
Author Manuscript
Figure 3: Glutamine and serine catabolism sustain the metabolic needs of collagen biosynthesis.
Author Manuscript
(A-B) Collagen concentration (media) of IL-6-activated PAAFs (A) and PH-PAAFs (B)
treated as indicated. (C-I) Schema of the experimental procedure (C). Representative image
(D and F) and quantification (E, G and I) of fibrillar collagen and vascular (PAEC and
PASMC) cell proliferation. (H) Matrix stiffness assessed by atomic force microscopy. Data
are represented by Tukey box-and-whisker plots. Median represents measures from n=3
matrices/group. In all panels **P<0.01; ***P<0.001; ANOVA with Bonferroni’s multiple
comparison test was used in (A-G, I); Kruskal-Wallis with Dunn’s post-hoc testing was
used in (H); data show mean ± s.d. Scale=20 μm. For all the panels, each dot represents a
Author Manuscript
biological replicates.
Author Manuscript
Author Manuscript
Author Manuscript
(A) Pathway enrichment analysis of genes with TEAD biding sites (ENCODE Project)
in their promoter regions. (B) Schematic of the collagen metabolism pathway. (C) TEAD
binding enrichment in the promoter region of indicated genes. (D) ChIP-qPCR confirmed
the presence of TEAD/YAP binding sites in the promoter regions of indicated genes.
Results are expressed as percentage of total input DNA prior to immunoprecipitation with
anti-YAP or anti-IgG control. (E-F) Heatmap with log2(fold change) range between −2 to 2
(E) and pathway enrichment analysis (F) of significantly (FDR<0.1; P<0.05) modulated
metabolites. (I) Matrix collagen concentration of PAAFs treated as indicated. (J-L) Schema
of the experimental procedure (J). Representative image (K) and quantification (L) of
fibrillar collagen and vascular (PAEC and PASMC) cell proliferation. Scale=20 μm. In all
panels *P<0.05; **P<0.01; ****P<0.0001; Mann-Whitney test was used in (D:SLC6A9-E;
SHMT1:E); two-tailed Student’s T-test was used in remaining panels of (D); ANOVA with
Bonferroni’s multiple comparison test was used in (I, L: Ki67 graphs); Kruskal-Wallis with
Dunn’s post-hoc testing was used in (L: area thresholded). Data show mean ± s.d. For all the
panels, each dot represents a biological replicates.
Author Manuscript
Author Manuscript
Author Manuscript
Figure 5: Genetic deletion of YAP and TAZ in vascular fibroblast prevents collagen biosynthesis
and ameliorates PH.
(A) Protocol for conditional YAP/TAZ deletion (cKO) in IL-6 transgenic mice. (B-C, E,
and H) Immunostaining and quantification for GLS1 (B), SHMT1 (C), collagen I (Col1;
E) and Ki67 (H) in pulmonary artery from YAP/TAZcKO mice or control mice (Ctrl).
(D) Collagen concentration (Total and fibrillar) of lungs from YAP/TAZcKO mice or Ctrl
mice. (F) Representative images and quantification of picrosirius red staining in pulmonary
artery from YAP/TAZcKO mice or Ctrl mice. (G) Pulmonary vascular stiffness assessed by
atomic force microscopy. Data are represented by Tukey box-and-whisker plots. Median
Author Manuscript
represents measures from n=3 mice/group. (I-K) Pulmonary arteriolar muscularization (I),
right ventricular systolic pressure (J), and right ventricular hypertrophy (Fulton index,
RV/LV+S; K) of YAP/TAZcKO mice or Ctrl mice. Scale=50 μm. In all panels, *P<0.05;
**P<0.01; ***P<0.001; ****P<0.0001; Mann-Whitney test was used in (F:Orthogonal;
G-I); two-tailed Student’s T-test was used in (B-E; F:Parallel; J-K). Data show mean ± s.e.m.
Panel B-C, E-F, and H-I: each dot represents a single artery from N=5 Control mice and N=6
cKO mice. Panel D and J-K each dot represents a single mouse
force microscopy. Data are represented by Tukey box-and-whisker plots. Median represents
Author Manuscript
measures from n=4 rats/group for (SHIN1 and CB-839+SHIN1) and n=7 rats/group for (Ctrl
and CB-839). (E) Immunostaining and quantification for Ki67 in pulmonary artery from
rats treated as indicated. (F-H) Pulmonary arteriolar muscularization (F), right ventricular
systolic pressure (G), and right ventricular hypertrophy (Fulton index, RV/LV+S; H) of rats
treated as indicated. Each dot represents a single rat. Scale=50 μm. In all panels *P<0.05;
**P<0.01; ***P<0.001; ****P<0.0001; ANOVA with Bonferroni’s multiple comparison test
was used in (B, C, E, H); Kruskal-Wallis with Dunn’s post-hoc testing was used in (D, F, G).
Data show mean ± s.e.m. In panels B-C, and E-H each dot represents a single rat
Author Manuscript
Author Manuscript
Author Manuscript
Figure 7: A glutamine- and serine-free diet prevents pulmonary vascular stiffening and PH.
Author Manuscript
(A) Rats received normal chow until 12 weeks of age; they were then transferred to either
a control diet (synthetic diet, containing glutamine and serine) or a matched diet lacking
glutamine and serine (Gln−/Ser− diet). Eighteen (18) days after diets were initiated, rats
were injected with monocrotaline. At Day 39, rats were euthanized and assessed for PH. (B)
Serum levels of glutamine, serine, and glucose were analyzed at serial time points. Mean
expression in control group was assigned a fold change of 1, to which relevant samples were
compared. (C) At D39, serum levels of the indicated metabolites were analyzed. Unpaired
two-tailed Student’s T-test and Mann-Whitney test. (D) Collagen concentration (total and
Author Manuscript
fibrillar) of lungs from rats treated as indicated. (E) Immunostaining and quantification
for collagen I (Col1) in pulmonary artery from rats treated as indicated. (F) Pulmonary
vascular stiffness assessed by atomic force microscopy. Data are represented by Tukey
box-and-whisker plots. Median represents measures from n=7 rats/group for (standard diet)
and n=4 rats/group for (synthetic diet and Gln−/Ser− diet). (G-H) Immunostaining (G)
and quantification (H) for Ki67 in pulmonary artery from rats treated as indicated. (I-K)
Pulmonary arteriolar muscularization (I), right ventricular systolic pressure (J), and right
ventricular hypertrophy (Fulton index, RV/LV+S; K) of rats treated as indicated. Each
dot represents a single rat. Scale=50 μm. In all panels, *P<0.05; **P<0.01; ***P<0.001;
****P<0.0001; Mann-Whitney test was used in (C:glutamine/glutamate; phenylalanine/
tyrosine; D:soluble/insoluble collagen); two-tailed Student’s T-test was used in remaining
panels of (C) as well as (D: collagen). ANOVA with Bonferroni’s multiple comparison test
Author Manuscript
was used in (E, G, H, I, J); Kruskal-Wallis with Dunn’s post-hoc testing was used in (F).
Data show mean ± s.e.m. In panels C-E and G-J each dot represents a single rat.
Author Manuscript
Author Manuscript
Antibodies
Rabbit Polyclonal anti-MLC2 Cell Signaling Technology Cat# 3672S; RRID: AB_10692513
Mouse Monoclonal anti-P-MLC2 Cell Signaling Technology Cat# 3675S; RRID: AB_2250969
Mouse Monoclonal anti-Hsp90 Santa Cruz Biotechnology Cat# sc-69703; RRID: AB_2121191
Rabbit Monoclonal anti-SHMT1 Cell Signaling Technology Cat# 80715; RRID: AB_2799957
Rabbit Monoclonal anti-YAP Cell Signaling Technology Cat# 14074; RRID: AB_2650491
Rabbit Polyclonal anti-YAP Santa Cruz Biotechnology Cat# sc-15407; RRID: AB_2273277
Rabbit Polyclonal anti-TAZ Santa Cruz Biotechnology Cat# sc-48805; RRID: AB_2216639
Author Manuscript
Peroxidase-AffiniPure Donkey Anti-Mouse IgG (H+L) Jackson ImmunoResearch Labs Cat# 715–035-150; RRID:
AB_2340770
Peroxidase-AffiniPure Donkey Anti-Rabbit IgG (H+L) Jackson ImmunoResearch Labs Cat# 711–035-152; RRID:
AB_10015282
Donkey anti-Goat IgG (H+L) Antibody, Alexa Fluor 488 Thermo Fisher Scientific Cat# A11055; RRID: AB_2534102
Author Manuscript
Donkey anti-Rabbit IgG (H+L) Antibody, Alexa Fluor 594 Thermo Fisher Scientific Cat# A21207; RRID: AB_141637
Donkey anti-Mouse IgG (H+L) Antibody, Alexa Fluor 647 Thermo Fisher Scientific Cat# A31571; RRID: AB_162542
Biological samples
Deposited data
Oligonucleotides
siRNA Negative control Horizon Discovery Cat#D-001810–10
Recombinant DNA
Human: pcDNA Flag Yap1 Levy et al., 2008 Addgene; Plasmid #18881
Other (Diets)
Author Manuscript
Deposited Data
Data S1 This paper
Author Manuscript
Author Manuscript