Dbi 230004

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

162 Diabetes Volume 73, February 2024

Molecular Insights From Multiomics Studies of


Physical Activity
Wei Wei,1,2 Steffen H. Raun,3 and Jonathan Z. Long 1,2,4,5,6
Diabetes 2024;73:162–168 | https://doi.org/10.2337/dbi23-0004

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


Physical activity confers systemic health benefits and such as muscle, adipose tissue, liver, and pancreas. In the
provides powerful protection against disease. There has past decade, advancements in multiomics technologies—
been tremendous interest in understanding the molecular including metabolomics, proteomics, phosphoproteomics,
effectors of exercise that mediate these physiologic lipidomics, single-cell RNA sequencing (scRNA-seq), and epi-
effects. The modern growth of multiomics technologies— genomics—have offered unparalleled opportunities to ex-
including metabolomics, proteomics, phosphoproteomics, tensively investigate the molecular changes associated with
lipidomics, single-cell RNA sequencing, and epigenomics— physical activity at an organism-wide scale (Fig. 1). Numer-
has provided unparalleled opportunities to systematically ous exercise “omics” data sets start to emerge (8,9). These
DIABETES SYMPOSIUM

investigate the molecular changes associated with physical


data sets provide a high-dimensional and detailed view of
activity on an organism-wide scale. Here, we discuss how
the molecular processes involved in exercise, further refining
multiomics technologies provide new insights into the sys-
our understanding of the molecular events related to this
temic effects of physical activity, including the integrative
complex physiological stimulus. Simultaneously, the vast
responses across organs as well as the molecules and
mechanisms mediating tissue communication during exer-
amount of data generated with these modern techniques
cise. We also highlight critical unanswered questions have raised new questions and opened up potential avenues
that can now be addressed using these high-dimensional for discovery regarding the molecules and molecular mecha-
tools and provide perspectives on fertile future research nisms underlying physical activity’s beneficial effects.
directions. In this article, we will focus on the latest develop-
ments using multiomics approaches to explore the mol-
Regular physical activity can help reduce the incidence of ecules of physical activity. We begin with a discussion
obesity and obesity-associated metabolic diseases (1–3). In of how multiomics technologies have provided new in-
contrast, physical inactivity has become increasingly com- sights into the systemic effects of physical activity, in-
mon in the U.S. population (4), representing a significant cluding the integrative responses across organs and the
modifiable risk factor for cardiovascular diseases, obesity, molecules and mechanisms mediating tissue communi-
diabetes, cancer, and overall mortality (5,6). Understand- cation during exercise. We also discuss multiomics studies
ing the molecules and pathways involved in physical activ- that revisit the classical exercise-responsive metabolic tis-
ity’s cardiometabolic benefits has garnered significant sues, including muscle and adipose, and how these studies
interest (7). This understanding serves as a crucial first have extended our understanding of the responses of
step toward developing future therapies that could poten- nonparenchymal cells, including immune cells, to physical
tially harness the health benefits of exercise. activity. Lastly, we share our perspectives on the future
Historically, molecular studies of physical activity have fo- directions for multiomics technologies as applied to exer-
cused on specific molecules in individual metabolic tissues, cise research.

1
Department of Pathology, Stanford University School of Medicine, Stanford, CA Corresponding authors: Wei Wei, wwbiomed@stanford.edu, and Jonathan Z.
2
Sarafan ChEM-H, Stanford University, Stanford, CA Long, jzlong@stanford.edu
3
Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Received 10 April 2023 and accepted 4 June 2023
Denmark
4 This article is part of a special article collection available at https://diabetesjournals
Stanford Diabetes Research Center, Stanford University School of Medicine,
.org/collection/1824/Diabetes-Symposium-2023.
Stanford, CA
5
Stanford Cardiovascular Institute, Stanford University School of Medicine, © 2024 by the American Diabetes Association. Readers may use this article
Stanford, CA as long as the work is properly cited, the use is educational and not for
6
Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA profit, and the work is not altered. More information is available at https://
www.diabetesjournals.org/journals/pages/license.
diabetesjournals.org/diabetes Wei, Raun, and Long 163

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


Figure 1—Overview of multiomics technologies for investigating the complex molecular response to physical activity.

MULTIOMICS OF INTEGRATIVE EXERCISE levels of heat shock proteins increased was notably influ-
RESPONSES ACROSS THE ENTIRE ORGANISM enced by both the duration of exercise training and the sex
One insight from multiomics data sets is the coordinated of the rats. For example, in male rats, an 8-week training
molecular responses across multiple organ systems in re- regimen resulted in a 16-fold increase in the abundance of
sponse to physical activity. These responses include spe- HSPA1B and HSPB1 proteins, whereas only a 2-fold in-
cific pathways that are broadly activated across multiple crease was observed after 1 week of training. Additionally,
tissues, as well as the increased correlation of metabolic the intensity of the exercise-induced heat shock response
profiles between anatomically segregated cell types and was less significant in female rats, as the levels of individ-
organs. These studies collectively suggest that one effect ual HSPs did not increase by more than fourfold even after
of physical activity is to reset and to coordinate molecular an 8-week exercise training program (8).
processes across multiple organ systems. Metabolomics data sets of exercise have also revealed
For instance, several different multiomics data sets have the rewiring of intertissue metabolic coordination by exer-
revealed a coordinated activation of the heat shock re- cise training (Fig. 2). Sato et al. (12) performed undirected
sponse of multiple cell types and organs after exercise condition-specific correlation networks on >1,000 metab-
training. Heat shock proteins (HSPs) are a family of an- olites detected across eight tissues after a single bout
cient proteins that act as molecular chaperones, with the of treadmill running in 3-month-old male mice. They
specific role of preserving cellular homeostasis in response showed that this exercise bout significantly increased the
to stress (10). Previously, Salo et al. (11) showed that a spe- correlation of metabolites across several organs, including
cific member of the HSP family, HSP72, is induced in spe- in the serum, liver, heart, hypothalamus, skeletal muscle,
cific tissues such as the liver and skeletal and cardiac brown adipose tissue, and epididymal and inguinal white
muscle after acute exercise. In combining transcriptomic adipose tissue. Sato et al. further suggested that this in-
and proteomic analyses for 6-month-old rats of both sexes creased metabolite correlation between organs might re-
after 8 weeks of treadmill training, the Molecular Trans- flect exercise-dependent shifts in fuel preference during
ducers of Physical Activity Consortium (MoTrPAC) consor- physical activity. For example, during exercise, an increase
tium found evidence of a consistent activation of the heat in the correlation of amino acids was observed between the
shock response across multiple organs—including the cor- heart-serum-liver, suggesting an increased amino acid fuel
tex, heart, lung, liver, kidney, subcutaneous white adipose coordination between these two organs. Similarly, exercise
tissue, and skeletal muscle (8). The degree to which the increased the correlation between carbohydrates in brown
164 Multiomics Studies of Physical Activity Diabetes Volume 73, February 2024

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


Figure 2—Highlighted discoveries related to physical activity enabled by multiomics technologies. Proteomics: using cell type–selective
plasma proteomics, Wei et al. (26) mapped the secretomes of 21 different cell types in mice after exercise training. Posttranslational modifica-
tions: exploring the phosphoproteomic landscape in skeletal muscle after three different exercise modalities, Blazev et al. (42) discovered a
novel AMPK substrate, C18ORF25. Metabolomics: Sato et al. (12) showed how the metabolomes of different tissues are altered after acute ex-
ercise. Li et al. (28) discovered a metabolite, Lac-Phe, that is induced by high-intensity exercise and is involved in appetite regulation. RNA se-
quencing: using RNA sequencing Sun et al. (13) showed that lifelong exercise training can maintain proper circadian rhythmicity of genes in
aging mice. Lipidomics: Stanford et al. (43) showed that acute exercise leads to the release of the lipokine 12,13-diHOME, which in turn alters
the whole-body metabolism. FC, fold change.

fat and lipids in inguinal white fat, suggesting that coordi- et al. (13) showed that a 12-month voluntary wheel running
nated carbohydrate/lipid metabolism serves as an axis for in- intervention in 16-month-old male mice restored crucial cir-
teradipose cross talk in exercise. Another surprising finding cadian rhythm genes, such as transcription factor genes
from the metabolite correlation network is that the hypo- Bmal1 and Dbp, to levels comparable with those seen in
thalamus becomes the correlation hub between organs after 2-month-old young mice (Fig. 2). This restoration occurred in
acute exercise. Some of the underlying correlation signals re- >30 of the 101 cell types identified in 14 major tissues. These
vealed metabolites involved in amino acid metabolism, with cell types include cells that reside in the central nervous sys-
a strong enrichment of a class of poorly studied metabolites, tem, such as neurons, astrocytes, and oligodendrocytes, as
acetylated amino acids, after the exercise perturbation (30% well as in the peripheral tissues, such as macrophages,
of the total correlated metabolites in amino acid metabolism) smooth muscle cells, and endothelial cells. Furthermore, these
compared with the sedentary condition (8%). For example, exercise-inducible changes in circadian clock genes were also
an increase in correlation of N-acetyl-leucine was observed observed in young mice. In another study involving 12-week-
between hypothalamus-serum-skeletal muscle, suggesting a old male mice exposed to 3 weeks of voluntary wheel running,
potential exercise-inducible metabolite shuttle between pe- an enriched circadian rhythm pathway was identified in mes-
ripheral tissues and the brain. enchymal stem cells (MSCs) across skeletal muscle and subcu-
Lastly, organism-wide multiomics has also revealed an taneous and visceral white adipose tissue. After exercise, the
intimate and much more widespread connection between gene Dbp was once again increased, regardless of the diet
exercise and circadian rhythms than previously recognized. composition, in both chow- and high-fat diet–fed mice (14).
Recent studies have shown that exercise affects the transcrip- Using ATAC-Seq (assay for transposase-accessible chromatin
tion levels of circadian clock genes across the body in an age-, with sequencing) to map differentially accessible regions of
training regime–, and sex-independent manner but an exercise the whole genome, the MoTrPAC consortium showed that
training frequency–dependent manner. Using scRNA-seq, Sun open chromatin regions were enriched with sequence
diabetesjournals.org/diabetes Wei, Raun, and Long 165

motifs recognized by key transcription factors in circadian treadmill (27). More examples of such non-muscle-derived
clock programs, namely BHLHE41, NPAS, and BMAL1. bioactive proteins can be found in a comprehensive review
This circadian clock gene change was sex independent, as by Chow et al. (17).
this regulation pattern was found in 6-month-old rats of Beyond circulating polypeptides, physical activity also dra-
both sexes subjected to 8-week treadmill training (15). All matically regulates the levels of circulating, bioactive metabo-
of these observations show that exercise resets central and lites. Targeted and untargeted metabolomics analyses have
peripheral clocks in multiple tissues. Given that global and now identified hundreds of plasma metabolite changes fol-
tissue-specific disruptions of circadian rhythm programs lowing physical activity (12,17,22,28,29). For example, ma-
dramatically impair exercise adaptations and benefits (16), late and succinate, two metabolite intermediates in classical
these studies add to the growing body of evidence suggest- glycolysis and TCA cycle pathways, are dramatically elevated
ing complex and direct molecular interactions between in blood plasma following exercise (22). These metabolites
physical activity and circadian clocks across the organism. can exert signaling functions, such as regulating muscle re-
modeling, cell cycle progression, brain function, and lipid oxi-

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


MULTIOMICS OF CIRCULATING MOLECULES dation (30–33). Additionally, untargeted metabolomics has
THAT MEDIATE ORGAN CROSS TALK been a powerful approach for uncovering novel bioactive me-
A long-standing hypothesis is that exercise induces the tabolites associated with physical activity. For example, in
production of circulating signaling molecules, also known our recent study we identified a lactate–amino acid conjugate,
as “exerkines” or “exercise factors,” that mediate tissue N-lactoyl-phenylalanine (Lac-Phe), to be robustly induced by
cross talk and function as molecular effectors of physical exercise across multiple species (28). Pharmacological
activity (17). The first evidence for such hormonal factors gain- and genetic loss-of-function studies demonstrate that
dates back to 1961, when blood transfusions from exer- Lac-Phe regulates feeding and obesity. Exercise-inducible
cised dogs were found to lower blood glucose levels in rest- Lac-Phe production is likely driven by the dramatically in-
ing dogs (18). For many years, research focused primarily creased circulating lactate during exercise and establishes an
on secreted polypeptides from skeletal muscle, which have unexpected role for lactate-derived metabolite in physical
also been referred to as “myokines.” However, recent mul- activity (Fig. 2). Another example is the brown adipose–
tiomics studies have expanded both the cell types of origins derived linoleic acid metabolite 12,13-diHOME, which pro-
and the types of molecules that are induced by physical motes skeletal muscle fatty acid uptake (34). The increase in
activity. circulating 12,13-diHOME immediately after a single bout
A growing number of studies are indicating that of exercise suggests that exercise-induced lipolysis may be
exercise-regulated bioactive plasma proteins originate from responsible for the elevated levels of this metabolite (Fig. 2).
multiple sources beyond muscle (19–26). For example, we These studies collectively suggest that key metabolite effec-
recently used a cell type–specific secretome tagging strategy tors of exercise may be commonly derived from the principal
to identify new exercise-inducible secreted proteins (Fig. 2). metabolic fuels (e.g., lactate or fatty acids) that are mobilized
Our findings revealed that two intracellular carboxylester- during physical activity.
ases expressed in the liver generated extracellular proteo-
forms capable of antiobesity, antidiabetes, and endurance- MULTIOMICS REVEALS RESPONSES OF
enhancing effects in mice (26). Additionally, in two distinct NONPARENCHYMAL CELLS TO EXERCISE
studies with use of untargeted proteomic analysis of blood Traditionally, studies of the exercise response in key meta-
plasma, the induction of a complement protein and a phos- bolic tissues, such as adipose tissue, muscle, and liver, have
pholipase in mice subjected to voluntary wheel running was largely focused on molecular and physiologic changes at
reported. These proteins are primarily expressed in the liver the whole organ level. Recent advancements in multiomics
and were observed to reduce brain inflammation and en- techniques, particularly in scRNA-seq, have enabled a high-
hance cognitive function, respectively (20,21). The findings resolution reexamination of individual cell types’ response
of these studies collectively suggest that the liver is an es- in these major exercise-responsive tissues. This has led to
sential contributor to the exerkine pools during exercise. an increasing appreciation of nonparenchymal cell involve-
Besides the liver, other tissues such as bone can also act as ment in the exercise response in adipose tissue and skeletal
sources of exercise-induced bioactive plasma proteins. For muscle.
instance, after a single bout of treadmill running in mice For example, it is well established that exercise training
and humans, osteocalcin, a bone-derived circulating hor- induces muscle hypertrophy and increased capillary density.
mone, was induced in circulation. Osteocalcin was found to However, there has not been a detailed understanding of
enhance glucose uptake and catabolism and increase fatty the cell types contributing to such remodeling. In analyzing
acid oxidation in skeletal muscle. Notably, in mice, whole- scRNA-seq data using pseudotime trajectory analysis in hu-
body loss of osteocalcin or skeletal muscle–specific deple- mans subjected to a single bout of cycling exercise, Lovric
tion of Gprc6a, the osteocalcin receptor, led to comparable et al. (35) demonstrated that the transcriptional features of
outcomes of impaired endurance capacity, as demonstrated Pax71 satellite cells shifted toward Tnni21 fast-twitch and
by a 20–30% reduction in running time and distance on a Tnni11 slow-twitch myogenic cells. The authors concluded
166 Multiomics Studies of Physical Activity Diabetes Volume 73, February 2024

that this satellite-to-myocyte transition explains the cellular the most robust secretome response across all of the cell
sources of increased density of muscle fiber after exercise. types studied in response to 1-week treadmill training in
This finding was further supported in another single-nuclei mice (26). These cells could be found in adipose tissue and
sequencing study focusing on chronic exercise training. muscle, as well as additional nonmetabolic tissues such as
Here, Wen et al. (36) showed that the number of Pax71/ the lung. Following exercise, Pdgfra1 MSCs released vari-
Myf51 and Pax71/Myf5 significantly increased up to ous molecules into the bloodstream, including TIMP3,
50-fold after 4 weeks of weighted wheel training in mice. which regulates extracellular remodeling; F13A, which is
They conclude that exercise training increases Pax71 satel- involved in coagulation; and LOXL1, which contributes to
lite self-renewal and maturation. Both of these studies un- fibrosis. These findings suggest that MSCs play a systemic
derscore the power of single-cell sequencing to define the regulatory role in the modulation of whole-body physiology
cellular basis and differentiation trajectories that underlie after exercise.
skeletal muscle adaptations to exercise.
scRNA-seq has also revealed increased immune cell re- FUTURE PERSPECTIVES

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


cruitment and immune pathway activation in muscle and The use of multiomics approaches for analyzing molecular
adipose tissues following exercise. For example, in human changes during physical activity has revealed a more com-
skeletal muscle, it was recently demonstrated that both the plex exercise landscape than previously understood. This
lymphocyte cell population (including T, B, and natural killer comprehensive and unbiased strategy has unveiled sur-
[NK] cells) and the monocyte cell population exhibited a sub- prising results of exercise-induced effects, including the
stantial increase, from 4 to 9% and 2% to 4%, respectively, dramatic exercise response of nonparenchymal cells, the
following repeated bouts of all-out cycling sprints (35). In a integrative response across tissues/organ systems, and
separate study of trained and untrained human subjects fol- the intricate regulation of multiorigin exerkines. Despite
lowing acute bouts of bilateral knee extension, a Pathway- the wealth of data that has been collected, as well as the
Level Information ExtractoR (PLIER) deconvolution analysis new insights obtained, many open questions remain. Be-
of microarray data showed an increased proportion of neu- low, we share our perspectives on four fertile and exciting
trophils, identified by marker genes such as S100A8 and future research directions that are now addressable using
S100A9, in the skeletal muscle of both (37,38). Lastly, single- these high-dimensional tools.
cell transcriptomic analysis of white and brown adipose tis-
sues in male rats undergoing an 8-week treadmill training THE COMPLEX ROLE OF LACTATE AND ITS
regimen revealed a significant increase in the number of T, POTENTIAL AS A PRIMARY DRIVER OF THE
B, and NK cells, as indicated by increased transcript abun- EXERCISE RESPONSE
dance of canonical immune cell markers and enriched im- Blood lactate levels are one of the most common blood meas-
mune pathways such as the B and T cell receptor signaling urements during exercise testing, especially in athletes. By
pathway, which was consistent with the findings in skeletal both abundance and magnitude, lactate is one of the most dra-
muscle (8). The cellular origins of the increased immune cell matically regulated molecules in exercise: its levels can be ele-
recruitment and activation in exercise, as well as the func- vated >10-fold to >20 mmol/L after vigorous sprinting.
tional consequences of perturbing this process for exercise Beyond its primary metabolic role as an interorgan metabolic
adaptations, remain unclear at this time. fuel during exercise (e.g., the “lactate shuttle”), more recent
MSCs in adipose tissue and muscle are another nonpar- studies have identified even more unexpected functions for
enchymal cell type that displays an unexpectedly strong re- lactate in the context of the response to physical activity. For
sponse to physical activity. These stromal cells have the instance, we recently showed that the role of lactate also
unique ability to self-renew and differentiate into various extends to lactate-derived metabolites such as Lac-Phe, an
cell types, depending on their location within the tissue. exercise-inducible signaling metabolite that suppresses food
Due to their low tissue abundance, e.g., <1% in adipose tis- intake and obesity (28). In addition, extracellular lactate can
sue (39), MSCs have not been previously studied in the con- directly induce the secretion of intracellular carboxylesterase
text of exercise. A recent study with use of single-cell proteins from hepatocytes, which function as bioactive circu-
transcriptomic analysis of skeletal muscle and subcutaneous lating plasma proteins that regulate energy metabolism (26).
and visceral adipose tissues highlighted a central role of That some of these roles have only been identified, or further
MSCs in tissue adaptation to 3 weeks of voluntary running expanded upon, in the past few years suggests that we are
in mice (14). Among 22 cell types identified within muscle only in the early stages of understanding the diversity of lac-
and fat, MSCs had the largest number of exercise-induced tate functions in exercise physiology. One possibility for
differentially expressed genes. Pathway enrichment analysis speculation is that lactate elevation functions as a key driver
of these genes revealed the downregulation of extracellular for numerous secondary downstream molecular events in
matrix remodeling, highlighting a previously unknown cellu- exercise. Such a hypothesis has not been rigorously tested
lar contributor to local tissue remodeling. Similarly, in our but may provide a unifying framework for understanding
own work using a cell type–selective secretome profiling and organizing the thousands of molecular changes that
approach (40,41), we showed that Pdgfra1 MSCs exhibited occur in response to physical activity. We propose future
diabetesjournals.org/diabetes Wei, Raun, and Long 167

studies should prioritize exploring other potential roles of development of new tools in this area, such as neutralizing
lactate in exercise biology, as well as a consideration of how nanobodies or engineered enzymes for manipulation of spe-
lactate might function as a coordinating signal between the cific metabolite pathways, may enable the key functional ex-
molecular responses to physical activity. periments that demonstrate physiologic roles for exercise-
inducible circulating effectors in metabolism and physiology.
BIDIRECTIONAL AND CELL TYPE–SPECIFIC
RESPONSES TO EXERCISE “EXERCISE AS MEDICINE”—AN UNREALIZED
Despite specific exercise responses being conserved across DREAM
many cell types—such as upregulated heat shock response A crucial and exciting future direction of exercise research
and changes in circadian clock programs—many other cel- involves developing “exercise mimetics” that can pharmaco-
lular responses are tissue and cell type specific and can be logically imitate a wide range of health benefits associated
opposite depending on the tissue/cell type. For example, with physical activity. Most past efforts toward such mole-
exercise training leads to the recruitment and activation

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


cules have primarily targeted pathways that improve exer-
of immune cells in both white adipose tissue and skeletal cise performance. For example, erythropoietin increases
muscle, while decreasing the number of immune cells in hematocrit and VO2max for endurance performance; simi-
the small intestine of rats (8). This contrast in regulation larly, testosterone and other androgen receptor agonists in-
patterns between these different tissues indicates that crease lean mass and strength. However, performance is not
the immune effects of exercise are specific to each tissue. health. Salutary effects such as reduced cardiovascular mor-
Additionally, our recent studies profiling secreted proteins tality, increased bone strength, and improved brain health
released by different cell types revealed bidirectional regu- have yet to be conferred by therapeutic capture of exercise-
lation of the same proteins in different cell types by exer- regulated molecules or pathways. As multiomics techniques
cise. For example, secretion of a superoxide dismutase advance and we acquire more knowledge about the non-
was suppressed from pancreatic b-cells but induced from muscle/nonperformance benefits, we may discover new
myocytes and Pdgfra1 MSCs in mice following 1 week of molecules and opportunities to truly translate the concept
treadmill training (26). Taken together, these studies of “exercise as medicine” into reality.
show that many cellular responses to exercise are depen-
dent on the specific cell types under study and include
SUMMARY
many cell types that have not classically been studied as
part of the exercise response. Therefore, higher-resolution Exercise induces profound molecular changes on an organism-
approaches that can be applied in a cell type–specific or wide scale. In previous studies investigators have focused on
organ-specific manner become increasingly important for characterizing individual exercise-regulated molecules and
understanding these bidirectional responses to physical specific organs such as muscle and fat. Recent technical ad-
activity, which may become masked in studying more vances in multiomics technologies have allowed for deep
“bulk” samples. Importantly, future studies in this area mapping of molecular changes across the entire organism
should also be focused on capturing unusual cell types for and at high resolution. These data sets have revealed thou-
an understanding of the diversity of cellular responses to sands of molecular changes that are dependent on the exer-
exercise beyond those of classical metabolic tissues. cise subject (such as species, age, and sex), exercise modality
(acute vs. chronic, voluntary vs. involuntary, intensity and
A GROWING NEED FOR LOSS-OF-FUNCTION frequency), and a variety of environmental factors (such
STUDIES OF EXERKINES timing of exercise). They have also provided important in-
The majority of studies on candidate, exercise-inducible cir- sights into exercise physiology that were previously not well
culating molecular effectors have relied heavily on gain- understood, such as the role of exercise to coordinate molec-
of-function experiments. There are limited data available ular processes across multiple organ systems, or the dynamic
from loss-of-function models. Consequently, whether candi- responses of nonparenchymal cells to physical activity. We
date molecules are truly necessary for a particular physio- anticipate that in providing high-resolution insights into
logic effect of physical activity has remained a largely physical activity, these multiomics technologies will provide
unanswered question. The lack of loss-of-function studies is new insights that serve as a foundation for therapeutic
due, in part, to technical challenges. For instance, interpreta- “capture” of the benefits of physical activity for human
tion of genetic manipulation of secreted proteins can be health.
complicated due to concurrent ablation of intracellular
pools, which may have additional or secondary functions. Acknowledgments. The authors thank members of the J.Z.L. and
Neutralizing antibodies are typically not readily available for K.J. Svensson (Stanford University) laboratories for helpful discussions. The
most circulating polypeptides. For exercise-inducible metab- authors apologize for not citing the many other excellent articles in this area
olites, robust methods for their genetic manipulation can be due to space limitations.
difficult due to multiple biosynthetic pathways and/or unde- Funding. This work was supported by the National Institutes of Health
fined molecular pathways for their efflux from cells. The (DK105203 and DK124265) and the Wu Tsai Human Performance Alliance.
168 Multiomics Studies of Physical Activity Diabetes Volume 73, February 2024

Duality of Interest. No potential conflicts of interest relevant to this arti- 21. Horowitz AM, Fan X, Bieri G, et al. Blood factors transfer beneficial
cle were reported. effects of exercise on neurogenesis and cognition to the aged brain. Science
Prior Presentation. Parts of this study were presented in abstract form 2020;369:167–173
at the 83rd Scientific Sessions of the American Diabetes Association, San Diego, 22. Contrepois K, Wu S, Moneghetti KJ, et al. Molecular choreography of
CA, 23–26 June 2023. acute exercise. Cell 2020;181:1112–1130.e16
23. Guseh JS, Churchill TW, Yeri A, et al. An expanded repertoire of intensity-
References dependent exercise-responsive plasma proteins tied to loci of human disease
1. Knowler WC, Barrett-Connor E, Fowler SE, et al.; Diabetes Prevention risk. Sci Rep 2020;10:10831
Program Research Group. Reduction in the incidence of type 2 diabetes with 24. Kuramoto K, Liang H, Hong JH, He C. Exercise-activated hepatic
lifestyle intervention or metformin. N Engl J Med 2002;346:393–403 autophagy via the FN1-a5b1 integrin pathway drives metabolic benefits of
2. Hambrecht R, Walther C, M€obius-Winkler S, et al. Percutaneous coronary exercise. Cell Metab 2023;35:620–632.e5
angioplasty compared with exercise training in patients with stable coronary 25. Mittenb€uhler MJ, Jedrychowski MP, Van Vranken JG, et al. Isolation of
artery disease: a randomized trial. Circulation 2004;109:1371–1378 extracellular fluids reveals novel secreted bioactive proteins from muscle and
3. Blair SN, Kohl HW 3rd, Paffenbarger RS Jr, Clark DG, Cooper KH, Gibbons fat tissues. Cell Metab 2023;35:535–549.e7

Downloaded from http://diabetesjournals.org/diabetes/article-pdf/73/2/162/745699/dbi230004.pdf by guest on 24 February 2024


LW. Physical fitness and all-cause mortality. A prospective study of healthy 26. Wei W, Riley NM, Lyu X, et al. Organism-wide, cell-type-specific secretome
men and women. JAMA 1989;262:2395–2401 mapping of exercise training in mice. Cell Metab 2023;35:1261–1279.e11
4. Yang L, Cao C, Kantor ED, et al. Trends in sedentary behavior among the 27. Mera P, Laue K, Ferron M, et al. Osteocalcin signaling in myofibers is
US population, 2001-2016. JAMA 2019;321:1587–1597 necessary and sufficient for optimum adaptation to exercise. Cell Metab
5. Lear SA, Hu W, Rangarajan S, et al. The effect of physical activity on 2016;23:1078–1092
mortality and cardiovascular disease in 130000 people from 17 high-income, 28. Li VL, He Y, Contrepois K, et al. An exercise-inducible metabolite that
middle-income, and low-income countries: the PURE study. Lancet 2017;390: suppresses feeding and obesity. Nature 2022;606:785–790
2643–2654 29. Lewis GD, Farrell L, Wood MJ, et al. Metabolic signatures of exercise in
6. Booth FW, Roberts CK, Thyfault JP, Ruegsegger GN, Toedebusch RG. Role human plasma. Sci Transl Med 2010;2:33ra37
of inactivity in chronic diseases: evolutionary insight and pathophysiological 30. Reddy A, Bozi LHM, Yaghi OK, et al. pH-gated succinate secretion regulates
mechanisms. Physiol Rev 2017;97:1351–1402 muscle remodeling in response to exercise. Cell 2020;183:62–75.e17
7. Fan W, Evans RM. Exercise mimetics: impact on health and performance. 31. Agudelo LZ, Femenıa T, Orhan F, et al. Skeletal muscle PGC-1a1
Cell Metab 2017;25:242–247 modulates kynurenine metabolism and mediates resilience to stress-induced
8. MoTrPAC Study Group, Amar D, Gay NR, Jean Beltran PM, et al. depression. Cell 2014;159:33–45
Temporal dynamics of the multi-omic response to endurance exercise training 32. Roberts LD, Bostr€om P, O’Sullivan JF, et al. b-Aminoisobutyric acid
across tissues. 23 September 2022 [preprint]. bioRxiv:2022.09.21.508770
induces browning of white fat and hepatic b-oxidation and is inversely
9. Sanford JA, Nogiec CD, Lindholm ME, et al.; Molecular Transducers of
correlated with cardiometabolic risk factors. Cell Metab 2014;19:96–108
Physical Activity Consortium. Molecular Transducers of Physical Activity
33. Liu W, Wang Y, Bozi LHM, et al. Lactate regulates cell cycle by
Consortium (MoTrPAC): mapping the dynamic responses to exercise. Cell
remodelling the anaphase promoting complex. Nature 2023;616:790–797
2020;181:1464–1474
34. Stanford KI, Lynes MD, Takahashi H, et al. 12,13-diHOME: an exercise-induced
10. Henstridge DC, Febbraio MA, Hargreaves M. Heat shock proteins and
lipokine that increases skeletal muscle fatty acid uptake [published correction
exercise adaptations. Our knowledge thus far and the road still ahead. J Appl
appears in Cell Metab 2018;27:1357]. Cell Metab 2018;27:1111–1120.e3
Physiol (1985) 2016;120:683–691
35. Lovric A, Rassolie A, Alam S, et al. Single-cell sequencing deconvolutes
11. Salo DC, Donovan CM, Davies KJ. HSP70 and other possible heat shock
cellular responses to exercise in human skeletal muscle. Commun Biol
or oxidative stress proteins are induced in skeletal muscle, heart, and liver
2022;5:1121
during exercise. Free Radic Biol Med 1991;11:239–246
36. Wen Y, Englund DA, Peck BD, Murach KA, McCarthy JJ, Peterson CA.
12. Sato S, Dyar KA, Treebak JT, et al. Atlas of exercise metabolism reveals time-
Myonuclear transcriptional dynamics in response to exercise following satellite
dependent signatures of metabolic homeostasis. Cell Metab 2022;34:329–345.e8
13. Sun S, Ma S, Cai Y, et al. A single-cell transcriptomic atlas of exercise- cell depletion. iScience 2021;24:102838
induced anti-inflammatory and geroprotective effects across the body. 37. Raue U, Trappe TA, Estrem ST, et al. Transcriptome signature of
Innovation (Camb) 2023;4:100380 resistance exercise adaptations: mixed muscle and fiber type specific profiles
14. Yang J, Vamvini M, Nigro P, et al. Single-cell dissection of the obesity- in young and old adults. J Appl Physiol (1985) 2012;112:1625–1636
exercise axis in adipose-muscle tissues implies a critical role for mesenchymal 38. Rubenstein AB, Smith GR, Raue U, et al. Single-cell transcriptional
stem cells. Cell Metab 2022;34:1578–1593.e6 profiles in human skeletal muscle. Sci Rep 2020;10:229
15. Smith GR, Zhao B, Lindholm ME, et al. Multiomic identification of key 39. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose
transcriptional regulatory programs during endurance exercise training. tissue: implications for cell-based therapies. Tissue Eng 2001;7:211–228
12 January 2023 [preprint]. bioRxiv:2023.2001.2010.523450 40. Wei W, Riley NM, Yang AC, et al. Cell type-selective secretome profiling
16. Gabriel BM, Zierath JR. Circadian rhythms and exercise - re-setting the in vivo. Nat Chem Biol 2021;17:326–334
clock in metabolic disease. Nat Rev Endocrinol 2019;15:197–206 41. Wei W, Riley NM, Lyu X, Bertozzi CR, Long JZ. Protocol for cell type-
17. Chow LS, Gerszten RE, Taylor JM, et al. Exerkines in health, resilience specific labeling, enrichment, and proteomic profiling of plasma proteins in
and disease. Nat Rev Endocrinol 2022;18:273–289 mice. STAR Protoc 2021;2:101014
18. Goldstein MS. Humoral nature of the hypoglycemic factor of muscular 42. Blazev R, Carl CS, Ng Y-K, et al. Phosphoproteomics of three exercise
work. Diabetes 1961;10:232–234 modalities identifies canonical signaling and C18ORF25 as an AMPK substrate
19. Robbins JM, Peterson B, Schranner D, et al. Human plasma proteomic regulating skeletal muscle function. Cell Metab 2022;34:1561–1577.e9
profiles indicative of cardiorespiratory fitness. Nat Metab 2021;3:786–797 43. Stanford KI, Lynes MD, Takahashi H, et al. 12,13-diHOME: an exercise-
20. De Miguel Z, Khoury N, Betley MJ, et al. Exercise plasma boosts memory induced lipokine that increases skeletal muscle fatty acid uptake. Cell Metab
and dampens brain inflammation via clusterin. Nature 2021;600:494–499 2018;27:1111–1120.e3

You might also like