Abstract
Emerging evidence has unveiled heterogeneity in phenotypic and
transcriptional alterations at the single-cell level during oxidative
stress and senescence. Despite the pivotal roles of cellular
metabolism, a comprehensive elucidation of metabolomic heterogeneity in
cells and its connection with cellular oxidative and senescent status
remains elusive. By integrating single-cell live imaging with mass
spectrometry (SCLIMS), we establish a cross-modality technique
capturing both metabolome and oxidative level in individual cells. The
SCLIMS demonstrates substantial metabolomic heterogeneity among cells
with diverse oxidative levels. Furthermore, the single-cell metabolome
predicted heterogeneous states of cells. Remarkably, the pre-existing
metabolomic heterogeneity determines the divergent cellular fate upon
oxidative insult. Supplementation of key metabolites screened by SCLIMS
resulted in a reduction in cellular oxidative levels and an extension
of C. elegans lifespan. Altogether, SCLIMS represents a potent tool for
integrative metabolomics and phenotypic profiling at the single-cell
level, offering innovative approaches to investigate metabolic
heterogeneity in cellular processes.
Subject terms: Metabolomics, Senescence, Metabolomics
__________________________________________________________________
Integrated analysis of metabolome and oxidative stress at single-cell
level is challenging. Here, the authors develop SCLIMS, enabling
simultaneous profiling of metabolome and oxidative stress levels and
discoveries of key metabolites regulating oxidative stress, senescence,
and healthy aging.
Introduction
With the advancements in single-cell analysis techniques, the
heterogeneity of single-cell genetics, proteomics, and metabolomics has
captivated the attention of scientists for decades. The phenotypic and
transcriptional heterogeneity of individual cells has been extensively
elucidated in diverse biological processes, encompassing development,
cancer, and aging. For instance, through the lens of single-cell
transcriptomic studies, developmental trajectories are meticulously
unraveled, unveiling the exquisite tapestry of single-cell
heterogeneity and sub-populations during embryonic
development^[46]1,[47]2. The genetic and epigenetic heterogeneity
exhibited by cancer cells^[48]3 poses significant challenges in the
field of cancer therapy^[49]4. Delving into the single-cell
heterogeneity of these cells illuminates promising avenues for
therapeutic targets and strategies^[50]5,[51]6. It has been reported
that geriatric organs are mosaics of cells with varying ages, thereby
highlighting the remarkable heterogeneity exhibited by individual cells
during the process of aging^[52]7. Another study uncovers the
heterogeneity of transcriptional landscape in aged ovaries where genes
undergo specific changes during aging that are exclusive to different
oocyte subtypes^[53]8. In essence, comprehending cellular heterogeneity
is paramount in unraveling the intricate cellular mechanisms that
underlie biological processes, including development, aging, and
diseases.
A groundbreaking advancement in single-cell research lies in the
seamless integration of multiple features or functions within
individual cells, known as cross-modality analysis. This includes
integrating single-cell omics with cellular function or phenotype,
resulting in a truly remarkable and sophisticated
approach^[54]9,[55]10. For instance, with the Patch-seq technique,
single-cell transcriptome and electric activities are combined, linking
transcriptome to cellular function^[56]11. In a study investigating the
exocytosis and excretion function of pancreatic β-cells in diabetes,
the Patch-seq technology was employed to establish a connection between
β-cell functionality, as represented by electrical activities, and gene
expression profiles. This innovative approach successfully identified
genes that are closely associated with β-cell dysfunction^[57]11.
Another study on extracellular vesicles (EV) links lipidomics and
proteomic with EV membrane function and crosstalk of discrete tissues
in different stages of COVID-19 infection^[58]12. The integrative
analysis of metabolism and transcriptome reveals a rare subpopulation
of hyperactive T cells and subtypes of monocyte which links to disease
severity in COVID-19, suggesting an association between multi-omics and
immune cell functionality^[59]13. The combination of single-cell RNAseq
and fluorescence-activated cell sorting (FACS) illustrates the relation
between gene expression and stem cell function represented by specific
surface markers, discovering key molecules associated with self-renewal
in stem cells^[60]14. Thus, the cross-modality analysis enables a more
comprehensive view of single-cell omics and a deeper understanding of
cellular function, shedding light on mechanisms underlying complicated
biological processes.
Metabolism serves as both a reflection and regulator of cellular
function^[61]15. Currently, most metabolomics research remains confined
to homogenate-level analysis involving the preparation of samples using
bulk tissue or cell suspension. Consequently, the enigmatic metabolic
heterogeneity exhibited by cells during biological processes and the
underlying mechanisms at the single-cell level remain shrouded in
ambiguity. Recently, our laboratory and others have pioneered the
development of cutting-edge single-cell mass spectrometry techniques,
enabling us to unveil the intricate metabolic heterogeneity at an
unprecedented resolution^[62]16–[63]18. These new techniques mainly
focused on the interpretation of the metabolome without the phenotype
of single cells, omitting cross-modality features important for the
better understanding of cellular metabolism and function. However, the
integration of metabolic heterogeneity with phenotypic heterogeneity,
such as distinct levels of oxidative or senescent states within
individual cells, remains a formidable technical challenge.
In the present study, we employ a combination of single-cell mass
spectrometry (SCMS) and live-cell imaging techniques to simultaneously
capture the metabolomic features and cellular identities of individual
cells, thereby establishing a link between the metabolome and cellular
status at a single-cell resolution. By utilizing this approach, we
design our study using a cellular model of oxidative stress-induced
senescence, establishing the correlation between single-cell metabolome
and their oxidative or senescent status. This technique unlocks
possibilities in the realm of cross-modality analysis, integrating
single-cell metabolome with fluorescent labeling techniques, thereby
paving the way for innovative discoveries at the single-cell level.
Results
Integration of live-cell imaging and single-cell mass spectrometry
To simultaneously capture the metabolome and phenotypic features of a
single cell, we combined single-cell live imaging with mass
spectrometry technique^[64]17,[65]18 (SCLIMS) and setup a
cross-modality analysis platform. We employed a cellular oxidative
stress (OS) model^[66]19–[67]24 which can be readily induced and
labeled with dichlorodihydrofluorescein diacetate (DCFDA), a widely
utilized live-cell probe for the detection of cellular
OS^[68]8,[69]25,[70]26. Briefly, HEK293T cells were incubated with
H[2]O[2] in culture medium at a final concentration of 80 μM for 1 h,
and the medium was then replaced by fresh medium. The cells were
allowed to recover for 48 h prior to subsequent testing. Cellular
viability remained uncompromised in the model and was deemed suitable
for SCMS analysis (Supplementary Fig. [71]1a). The OS level was
evaluated through DCFDA staining^[72]8,[73]25,[74]26 (Supplementary
Fig. [75]1b, c). Initially, the cells were incubated with DCFDA for a
duration of 25 min and then imaged using a microscope. Subsequently,
cellular sampling was performed via patch clamp technique utilizing
micro-pipettes, followed by SCMS analysis. Finally, the fluorescent
intensity was calculated and paired with metabolomic features in single
cells, yielding a pairwise dataset of metabolome and oxidative levels
(Fig. [76]1a).
Fig. 1. The cross-modality analysis platform integrating single-cell
metabolome and cellular phenotype.
[77]Fig. 1
[78]Open in a new tab
a A workflow and experimental setup of the cross-modality analysis.
Cells were first labeled with DCFDA and photographed with a fluorescent
microscope, followed by sampling and single-cell MS analysis. The
oxidative levels were reflected by DCFDA fluorescent intensity and the
metabolic information was acquired by SCMS. b Heatmap showing relative
abundance of representative metabolites corresponding to single-cell
DCFDA intensity. DCFDA intensity was indicated by color: dark green,
relative high intensity; light green, relative low intensity.
Metabolite abundance was represented by color: red, relative high
abundance; blue, relative low abundance. c–e PCA score plot (c), UMAP
analysis (d), and tSNE analysis (e) showing no significant difference
in metabolome of DCFDA incubated (n = 257, pink) and non-incubated
cells (n = 325, blue). f Correlation heatmap illustrating Pearson’s
correlation coefficient (r) between metabolites in non-incubated (left,
n = 325) and DCFDA incubated (right, n = 257) cells. Glutamate was
correlated with GABA and glutamine (inset). Two-sided Pearson’s
correlation analysis was performed. P values were not adjusted. For
(c–e), Source data are provided as Source Data files.
In m/z ranging from 67 to 1000, a total of more than 500 ion signals,
with signal-to-noise ratio greater than 3 (refs. ^[79]27,[80]28) and
detected at a frequency greater than 20% in all single cells were used
in subsequent analysis. Among these signals, 162 matched annotations in
the HMDB database through MS spectra. The annotated metabolites
underwent further scrutiny via MS/MS, resulting in confirmation of 83
metabolites that were subsequently employed for pathway enrichment
analysis (Supplementary Data File [81]1). Fluorescent intensities of
sampled cells were calculated and matched to every single cell. The
heatmap illustrated the metabolite abundance and corresponding DCFDA
intensity in single cells, showing the gradual alteration of metabolism
as DCFDA intensity differs (Fig. [82]1b). A variety of metabolites,
exhibiting diverse changes in response to DCFDA intensification,
including glutathione (GSH), phosphocreatine, hypotaurine and adenosine
triphosphate (ATP), were unearthed, suggesting a profound impact of OS
on cellular metabolism.
To rule out the possibility that DCFDA incubation may disturb cellular
metabolism, we analyzed the metabolome of the cells treated with or
without DCFDA by using analysis methods including Principal component
analysis (PCA), uniform manifold approximation and projection (UMAP),
and t-distributed stochastic neighbor embedding (t-SNE). As a result,
no significant difference in cellular metabolome between DCFDA
incubated and non-incubated cells was observed (Fig. [83]1c-e).
Consistently, the abundance of 31 common metabolites including GSH,
ATP, adenosine diphosphate (ADP), glutamate, glutamine, creatine,
oxidized glutathione (GSSG), phosphorylcholine, and phosphoserine were
almost identical in DCFDA incubated and non-incubated cells
(Supplementary Fig. [84]1d). We further performed a correlation
analysis of metabolites in both DCFDA incubated and non-incubated
cells, revealing the similarity in the correlations among metabolites.
As shown in the heatmap, the correlation coefficients between
metabolites remain virtually indistinguishable in both DCFDA incubated
and non-incubated cells (Fig. [85]1f), suggesting that the metabolic
landscape was not significantly altered by DCFDA incubation. For
instance, the correlation and conversion of metabolites in the
classical glutamine-glutamate-GABA metabolic pathway remained
unaffected by the incubation of DCFDA, as evidenced by a robust
association between glutamate and GABA, as well as between glutamine
and glutamate, observed in both DCFDA-incubated and non-incubated
cells. The difference of correlation coefficients (r) between
non-incubated and DCFDA incubated cells was calculated and visually
represented using a heatmap (Supplementary Fig. [86]1e), illustrating
minimal divergence in the correlations of metabolites within the
non-incubated and DCFDA incubated cells. Altogether, the cross-modality
analysis platform successfully integrated single-cell metabolome and
cellular phenotype with high stability and reliability, while
preserving cellular viability and metabolism.
The SCLIMS reveals correlation between cellular metabolism and oxidative
levels
The Multi-modal properties of SCLIMS facilitate its capacity to explore
the link between cellular metabolism and their OS levels, by means of
conducting correlation analysis between single-cell metabolomics data
and DCFDA intensity in each cell (Fig. [87]2a). The metabolomics data
was confirmed with no obvious batch effect by PCA and HCA
(Supplementary Fig. [88]2a, b). For each cell, the DCFDA intensity and
the abundance of metabolites were concomitantly collected. Then the
correlation between DCFDA intensity and metabolite abundance was
analyzed, with the calculation of Pearson’s r coefficient to assess the
strength of the correlation. For example, the intracellular GSH
abundance exhibited an inverse correlation with the DCFDA intensity in
the cells (Fig. [89]2a). The consistency with previous studies, which
highlight the pivotal role of GSH as a crucial metabolite in
maintaining redox balance^[90]29,[91]30, further confirms our method’s
reliability.
Fig. 2. The correlation analysis of intracellular metabolome and cellular OS
level.
[92]Fig. 2
[93]Open in a new tab
a The diagram illustrates the pairing of metabolic data and OS levels,
as well as the subsequent correlation analysis. The DCFDA intensity
indicated the specific level of OS in each cell, while single-cell MS
was employed to determine the abundance of metabolites. Here, the
metabolite GSH in Cells 1–4 was used as an illustrative example. Data
from Cell 1–4 was normalized to Cell 1. The vectors for x and y were
constructed based on single-cell OS levels and metabolite abundances,
followed by calculation of Pearson’s coefficient (r) and significance
(P). In correlation analysis, the data was z scored. Scale bar, 15 μm.
b Metabolites that were inversely and positively correlated with the
level of OS. Metabolites are shown as dots by color: red, positively
correlated (P < 0.05); blue, inversely correlated (P < 0.05). The
representative metabolites are annotated adjacent to the corresponding
dots. P values were not adjusted. Two-sided Pearson’s correlation
analysis was performed. c–h The correlation analysis between the scaled
levels of OS and the scaled intensities of representative metabolites
including ATP (c), PCr (d), UTP (e), GTP (f), Hypt (g), and energy
charge (h). The data were standardized by calculating z score. P values
were not adjusted. Two-sided Pearson’s correlation analysis was
performed. i The metabolic pathways enriched by the MSEA analysis of
metabolites exhibiting a reverse correlation with OS levels. Only
metabolites with MS/MS confirmation were included in the analysis. Only
pathways with P < 0.05 in the MSEA analysis were included. P values
were not adjusted. Related metabolic processes are annotated on the
left. Dot size represents enrichment ratio, and dot color represents
significance of the enrichment (P value). Yellow, relatively high
significance; Blue, relatively low significance. n = 190 cells in (a)
and (c–h). For (a–h), Source data are provided as Source Data files.
ATP adenosine triphosphate, PCr phosphocreatine, UTP uridine
triphosphate, GTP guanosine triphosphate, Hypt hypotaurine, GSH
glutathione, CTP cytosine triphosphate, O-PE O-phosphoethanolamine,
G-3-P glycerol 3-phosphate, AMP adenosine monophosphate.
Among the whole metabolome, we observed a total of 254 metabolites
significantly correlated with OS level (P < 0.05). The majority (61.4%)
of the metabolites correlated with OS level (P < 0.05) exhibited an
inverse correlation, which is nearly double the number of metabolites
that showed a positive correlation with cellular OS. This suggests that
the downregulation of multiple metabolites may serve as a crucial
hallmark of OS (Fig. [94]2b). For instance, the abundance of key
metabolites associated with energy metabolism, such as ATP
(Fig. [95]2c) and phosphocreatine (Fig. [96]2d), exhibited a
progressive decline as OS level increased. The linear downregulation
was also observed in other high energy compounds such as uridine
triphosphate (UTP) and guanosine triphosphate (GTP) (Fig. [97]2e, f).
The abundance of hypotaurine (Hypt), which generates NADH as a
by-product upon conversion to taurine^[98]31 and plays crucial roles in
redox homeostasis and DNA protection^[99]32, gradually declines during
OS (Fig. [100]2g). The energy metabolism is composed of numerous
metabolites and is detrimentally affected by OS^[101]33. Single-cell
energy charge, a metabolic parameter used to assess cellular energy
supply by measuring ATP, ADP and adenosine monophosphate (AMP) levels,
reflects the energy homeostasis within a cell^[102]34. The inverse
correlation observed between the single-cell energy charge and the
cellular OS level (Fig. [103]2h) indeed suggests an association between
OS and dysfunctionality in energy metabolism.
By performing the metabolite set enrichment analysis (MSEA)^[104]35
using the metabolites inversely correlated with OS level, we found a
variety of metabolic pathways related to different biological processes
were disturbed in OS (Fig. [105]2i), such as the mitochondrial and
energy metabolism including “Mitochondrial Electron Transport Chain”
and “Citric Acid Cycle”, and the redox metabolism including
“Glutathione Metabolism Pathway”. These findings were in consistence
with previous reports observing the disruption of glutathione
metabolism^[106]36,[107]37 and energy metabolism^[108]33 during OS.
Lipid metabolism has been reported to be altered in OS^[109]38,[110]39.
This was also verified by our finding of the downregulated pathways
such as “Phosphatidylethanolamine Biosynthesis”, “Phosphatidylcholine
Biosynthesis”, and “Sphingolipid metabolism”. The downregulation of
purine and pyrimidine metabolic pathways revealed by the SCLIMS was
also supported by previous studies reporting a depletion of purine and
pyrimidine nucleotide during OS^[111]40. The SCLIMS also revealed an
interference of vitamin metabolic pathways including “Thiamine
Metabolism Pathway” and “Riboflavin Metabolism Pathway” during OS,
which is in line with previous studies illustrating the crucial role of
thiamine and riboflavin in regulating OS^[112]41,[113]42. The
consistency of these discoveries by SCLIMS and previous reports again
suggests the reliability of the technique.
The SCLIMS additionally detected numerous previously undisclosed
metabolic alterations during OS. For instance, pathways related to
amino acids metabolism such as “glutamate metabolism”, “alanine
metabolism”, “arginine and proline metabolism” were all disturbed in
the cells with OS. Moreover, pathways related to sugar and derivatives
metabolism such as “Amino sugar metabolism”, “Fructose and mannose
degradation”, “Lactose synthesis”, “Lactose degradation”, and
“Nucleotide sugars metabolism” were also discovered to be downregulated
in OS. Other pathways like “threonine and 2-oxobutanoate degradation”
were also found to be disturbed in OS (Fig. [114]2i). The discoveries
made by SCLIMS have shed light on the pivotal role of metabolism in
cellular oxidative stress, unveiling a diverse array of metabolic
pathways that undergo significant alterations (P < 0.05). These
findings indicate that the mechanisms governing protein synthesis and
degradation, glycosylation reactions, and energy metabolism may undergo
drastic transformations under OS.
The SCLIMS technique was also employed for multi-modal analysis in
MEFs. Firstly, the metabolomics data were validated for the absence of
any noticeable batch effect through PCA and HCA (Supplementary
Fig. [115]2c, d). In MEFs, the key metabolites found to be inversely
correlated with OS levels in HEK cells, including GSH, O-PE, CTP, ATP,
UTP, Hypt, GTP, and PCr, were similarly observed to exhibit reverse
correlation with single-cell OS levels (Supplementary Fig. [116]3a).
Additionally, MSEA analysis was conducted using these metabolites
exhibiting inverse correlation with OS levels to unravel the metabolic
pathways involved in oxidative stress of MEFs. Similar to the results
of HEK cells, a variety of metabolic pathways were enriched
(Supplementary Fig. [117]3b). For example, “Pyruvate metabolism”,
“Citric acid cycle”, “Beta oxidation of very long chain fatty acids”,
and “Mitochondrial electron transport chain”, in mitochondria and
energy metabolism were also involved in the OS of MEFs. Lipid
metabolism, including “Phosphatidylcholine Biosynthesis”,
“Phosphatidylethanolamine Biosynthesis”, “Sphingolipid metabolism”,
were altered during OS in MEFs as well. The downregulation of purine
and pyrimidine metabolic pathways were also discovered in MEFs under
OS. Similar to the interruption of vitamin metabolism in HEK cells
under OS, “Pantothenate and CoA Biosynthesis”, “Thiamine Metabolism
Pathway” were also discovered to be downregulated in OS-stressed MEFs.
The outcomes in other pathways also exhibited a resemblance to those
observed in HEK cells. These results confirmed the robustness of the
multi-modal analysis of SCLIMS across diverse cellular phenotypes.
Taken together, revealing a profound interplay, the SCLIMS-based
cross-modality analysis has unveiled strong connections between
single-cell metabolism and OS, thereby highlighting the paramount
importance of integrating metabolome and cellular phenotype for
comprehensive insights across different cell types.
Cell types identified by the SCLIMS exhibit divergent OS levels
Cluster analysis was conducted using k-medoids
algorithm^[118]43,[119]44 based on the single-cell metabolome acquired
from the SCLIMS. As a result, cells were clustered into six subtypes
(C1–C6) with distinct metabolic features according to their metabolome
profiles, highlighting the diversity of metabolic characteristics
within these cells (Fig. [120]3a). Notably, the six subtypes of cells
displayed distinct levels of OS as indicated by varying DCFDA
intensities (Fig. [121]3b). We then performed a pseudotime analysis
using single-cell metabolomics data and generated a trajectory of the
six subtypes. The trajectory originated from subtype C1, which
exhibited the lowest level of OS, and gradually progressed towards
other subtypes. Notably, three distinct branches were observed that led
to C2/3 (representing a low oxidative level), C4 (representing a medium
oxidative level), and C5/6 (representing a high oxidative level),
respectively (Fig. [122]3c). These findings suggest a metabolism-guided
stepwise progression of cellular OS.
Fig. 3. Subtype-specific metabolic signatures and dynamic change of
metabolome revealed by cross-modality analysis.
[123]Fig. 3
[124]Open in a new tab
a UMAP visualization of six cellular subtypes based on the single-cell
metabolome. Each point represents a single cell, color represents
different subtypes. n = 55, 34, 14, 41, 29, and 17 for C1, C2, C3, C4,
C5, and C6 respectively. b Average OS levels (indicated by the
normalized DCFDA fluorescence intensity) of the six metabolic
subpopulations. n = 55, 34, 14, 41, 29, and 17 for C1, C2, C3, C4, C5,
and C6, respectively. F(5, 184) = 5.880, P = 4.57e−5 in One-way ANOVA
(labeled in the plot). Data was normalized to values of the respective
control (Cluster C1). Data is presented as mean ± s.e.m. Color
represents different subtypes and each dot represents a cell. c
Single-cell trajectory of pseudotime analysis showing temporal
progression of cell subtypes originating from C1 and gradually
transitioning towards C2/3, C4, and C5/6. Each subtype is represented
by a distinct color. For (a–c), Blue: C1; yellow: C2; purple: C3;
green: C4; brown: C5; red: C6. d Heatmap of characteristic metabolites
and their corresponding relative intensities in each subtype. Color
indicates z scores of metabolite abundance. Red: relatively high
abundance; blue: relatively low abundance. e Representative metabolic
pathways significantly (P < 0.05) enriched in Cluster C1 and C6 through
the MSEA analysis. Only metabolites with MS/MS confirmation were
included in the analysis. Dot size represents enrichment ratio, while
dot color indicates significance (−log[10] P value) of the enrichment.
Red: relatively high significance; blue: relatively low significance. P
values were not adjusted. For (a–c), Source data are provided as Source
Data files. Data was collected from at least three biological
replicates.
We subsequently conducted a more detailed analysis of the six metabolic
subtypes by utilizing a Wilcox rank sum test to compare metabolite
abundance between each cluster and other clusters. Each subtype was
characterized with specific metabolic markers as shown in the heatmap
(Fig. [125]3d). For instance, cells of Cluster C1 exhibited enrichment
of energy rich phosphate compounds such as ATP, GTP, UTP,
phosphocreatine along with antioxidants including GSH; cells of Cluster
C2 were enriched with dimethyldithiophosphate and pyrophosphate; cells
of Cluster C3 showed an abundance of catecholamine derivatives such as
homovanillic acid and sugar derivatives such as deoxyribose
5-phosphate; cells of Cluster C4 were enriched with intermediates of
nucleotide metabolism such as guanine monophosphate (GMP), along with
acetylated compounds such as N-acetylneuraminic acid and
UDP-N-acetylglucosamine; cells of Cluster C5 were enriched with amino
acids such as proline, threonine, asparagine, alanine and taurine,
along with glucose and ribitol; Nucleotide monophosphates, including
AMP, cytidine monophosphate (CMP), and uridine monophosphate (UMP), as
well as intermediates in the glycolysis pathway (i.e. glyceraldehyde
3-phosphate and fructose 1,6-bisphosphate), were found to be enriched
in cells belonging to Cluster C6.
Next, we conducted a comparison of the metabolome at the single-cell
level between Cluster C6 (with the most oxidative level) and Cluster C1
(with the least oxidative level). A total of 148 metabolites were
downregulated and only 64 metabolites were upregulated from C1 cells to
C6 cells (Supplementary Fig. [126]4a). To investigate the alteration of
the metabolic process in cellular OS, we performed the MSEA analysis of
the characteristic metabolites enriched in C1 and C6 cells
(Fig. [127]3e), and other clusters of cells (Supplementary
Fig. [128]5). Specifically, pathways related to lipid metabolism such
as “Phosphatidylethanolamine Biosynthesis”, “Phosphatidylcholine
Biosynthesis”, “Sphingolipid Metabolism”, and “Phosphatidylinositol
Phosphate Metabolism” were less enriched or completely depleted in C6
cells, consistent with previous reports^[129]38,[130]39. The pathways
associated with mitochondrial function and redox balance, such as
“Citric Acid Cycle”, “Nicotinate and nicotinamide metabolism” and
“Glutathione metabolism” were also found to be depleted or less
enriched in cells of C6, thus confirming previous reports on the
disruption of glutathione metabolism^[131]36,[132]37 and energy
metabolism^[133]33 under OS. The pathways related to metabolism of
sulfinic acids and organosulfonic acids, such as hypotaurine and
taurine metabolism, were depleted in C6 cells. This was supported by
studies illustrating the antioxidant effect of these
metabolites^[134]45,[135]46. In addition, the glycine and serine
metabolism exhibited greater enrichment in C6 cells, which may be a
compensatory response of cells under OS as glycine and serine
metabolism are reported to attenuate OS in C. elegans^[136]47. The
nucleotide metabolism pathways, such as “Purine metabolism” and
“Pyrimidine metabolism”, were depleted in C6 cells. The metabolism of
vitamins, including “Pantothenate and CoA metabolism”, “Thiamine
metabolism”, and “Folate metabolism”, exhibited reduced enrichment or
depletion in C6 cells. The alteration of nucleotide and vitamin
metabolism in OS were well described by previous
researches^[137]40–[138]42. Other pathways such as “Plasmalogen
Synthesis” was enriched exclusively in the cells of C6 (Fig. [139]3e).
Plasmalogen synthesis is reported to enhance the resistance to OS in E.
Coli.^[140]48, which could be served as a protective response in cells
under OS. Together, the convergence of these consistent findings
between the SCLIMS and the existing literature further substantiates
the dependability and steadfastness of this technique.
Moreover, the SCLIMS has also unearthed previously unreported perturbed
pathways during OS. For instance, several metabolic pathways, such as
“Fructose and mannose degradation”, “Amino sugar metabolism”,
“Nucleotide sugars metabolism” along with amino acids metabolism
including “alanine metabolism”, “glutamate metabolism”, and “arginine
and proline metabolism”, were identified to be implicated in OS, which
has not been elucidated in previous studies. Another finding gleaned
from the SCLIMS is the transition from the “Malate-Aspartate Shuttle”
pathway to the “Glycerol Phosphate Shuttle” pathway, observed between
C1 and C6, implying a reduced efficiency in ATP generation during OS.
The metabolic heterogeneity of MEFs under OS was meticulously
investigated using SCLIMS, revealing a parallel to HEK293T cells where
six distinct subtypes (M1–M6) with specific metabolic characteristics
were identified (Supplementary Fig. [141]6a). These subtypes in MEFs
also exhibited varying levels of OS (Supplementary Fig. [142]6b).
Furthermore, pseudotime analysis utilizing single-cell metabolic
features unveiled a trajectory of the six subtypes originating from M1
(with the lowest level of OS) and progressing towards M3 (with moderate
OS), followed by M5 and finally converging at M4/6 (with higher levels
of OS) (Supplementary Fig. [143]6c). Notably, a metabolism-guided
progression of cellular oxidative stress was similarly observed in
MEFs.
The metabolite abundance was also analyzed between each cluster and
other clusters, revealing subtype-specific metabolic markers
(Supplementary Fig. [144]6d). In comparison to HEK293T cells, MEFs
exhibited strikingly similar metabolic characteristics. Similar to C1
in HEK293T cells, M1 in MEFs shared crucial metabolites such as Hypt,
PCr, O-PE, ATP, GTP, UTP, and GSH. Both C1 and M1 displayed the lowest
level of OS. A comparison of the metabolites in M6 (with the highest OS
level) and M1 (with the lowest OS level) yielded a result akin to that
observed in HEK293T cells. A total of 195 metabolites were
downregulated, while 134 metabolites were upregulated from M1 to M6
(Supplementary Fig. [145]4b). The metabolic pathways enriched in M1 and
M6 were also analyzed using MSEA and compared (Supplementary
Fig. [146]6e). Consistent with the findings in C1 and C6 of HEK293T
cells, common lipid metabolic pathways such as
“Phosphatidylethanolamine Biosynthesis”, “Phosphatidylcholine
Biosynthesis”, and “Sphingolipid Metabolism” exhibited depletion in M6
MEFs characterized by the highest level of OS. Additionally, key
pathways involved in mitochondrial function and redox balance, namely
“Citric Acid Cycle” and “Glutathione metabolism”, were also found to be
depleted in M6 MEFs. Furthermore, several other pathways including
“taurine and hypotaurine metabolism”, “alanine metabolism”, “glutamate
metabolism”, and “arginine and proline metabolism” were identified as
being implicated specifically in the OS subtype of MEFs at stage M6.
Similarly, nucleotide metabolism pathways including “Purine metabolism”
and “Pyrimidine metabolism” showed depletion or less significant
enrichment in the same group of cells. Moreover, there was a depletion
observed in the vitamin-related metabolic processes such as
“Pantothenate and CoA metabolism”, “Thiamine metabolism”, and “Folate
metabolism” within the context of OS progression among these specific
type of cells. The obtained results exhibited consistency with those
observed in HEK293T cells, as well as previous studies that have
dissected cellular OS metabolism mentioned above. Consequently, SCLIMS
has been demonstrated to possess robustness in analyzing metabolic
alterations at the single-cell level.
Taken collectively, the SCLIMS not only validate metabolic changes
observed in previous studies but also unveil metabolic modifications
associated with OS and metabolic heterogeneity within individual cells.
These findings emphasize the reliability and universality of this
technique, establishing it as a powerful tool for comprehensive
analysis at the single-cell level.
The SCLIMS reveals capability of the single-cell metabolome in predicting
cellular OS status
Although the metabolic alterations and heterogeneity of cells under OS
were explored in detail, the strength of the link between metabolism
and cellular phenotype is not fully established. Machine learning is
utilized to ascertain whether the metabolic profile of individual cells
can accurately predict heterogeneous subtypes with distinct OS levels.
We employed discriminant analysis algorithms to train classification
models using single-cell metabolic features aiming to distinguish the 6
subtypes with specific OS levels identified in the above-mentioned
clustering analysis (Fig. [147]4a). The data was randomly divided into
independent training and testing datasets, with 2/3 of the original
data composing the former and 1/3 of it composing the latter. Both
datasets included all m/z signals meeting our criteria (S/N > 3 and
detected in greater than 20% single cells) and no variable selection is
performed before the model was trained. The model was trained on the
training dataset to acquire the ability to classify metabolic subtypes
based on full features of the single-cell metabolome, and its
performance was assessed using the testing dataset. The performance of
the model was evaluated using receiver operating characteristic (ROC)
curve and confusion matrix. In multiclassification, the ROC curve had
an average area under curve (AUC) of 0.98 (Fig. [148]4b). In cluster
prediction, the model achieved an accuracy ranging from 77.8% to 100%
(Fig. [149]4c). These results demonstrate that the single-cell
metabolic profiles can directly predict metabolic subtypes.
Fig. 4. Machine learning-guided prediction of OS levels based on single-cell
metabolome.
[150]Fig. 4
[151]Open in a new tab
a Flowchart of classification analysis with machine learning. The
training and testing dataset were randomly assigned according to a
ratio of 2:1. The model was trained and built with the training dataset
with 5-fold cross validation. The testing dataset was held out and used
for the evaluation of model accuracy independently. The performance of
the model was evaluated with ROC curve and confusion matrix. b ROC
curve of model testing. AUC for each cluster was determined separately
by the classification model. Higher AUC value indicates a better
performance of the model in predicting the clusters. The average AUC
represented an overall performance of the model. The color represents
classification of a certain subtype. Blue: C1; yellow: C2; purple: C3;
brown: C4; green: C5; red: C6. c Confusion matrix of model testing,
illustrating the distribution of errors in multi-class prediction. The
average accuracy was used to evaluate the overall performance of the
model. The color depth indicates the proportion of correct (blue) and
incorrect (red) predictions, as displayed in the bar chart. d Flowchart
of building a regression model with machine learning. The training and
testing datasets were randomly assigned according to a ratio of 2:1.
The model was trained and built with the training dataset with 5-fold
cross validation. The testing dataset was held out and used for the
evaluation of the model independently. e The correlation of real values
and values predicted by the regression model (n = 61). Dash line
represents the perfect fit (predicted values = real values). The model
performance was evaluated by the correlation coefficient (r) and P
value (P). Two-sided Pearson’s correlation analysis was performed.
P = 1.45e−20. P value was not adjusted. For b and e, Source data are
provided as Source Data files. AUC area under curve.
The classification model is capable of predicting metabolic subtypes
rather than precise levels of OS. We further trained a regression
model^[152]49,[153]50 based on neuronal network algorithms
(Fig. [154]4d), which may enable direct prediction of single-cell OS
levels utilizing metabolic features. Similarly, the data was
independently and randomly divided into training and testing datasets,
with 2/3 of the original data comprising the former and 1/3 comprising
the latter. All m/z signals meeting our criteria (S/N > 3 and detected
in greater than 20% single cells) were included in building the
regression model without variable selection. The model was trained on
the training dataset to predict single-cell OS levels, as manifested by
DCFDA intensity, based on single-cell metabolic features. The
performance of the model was evaluated with the testing dataset, and
the predicted values were plotted against real values (Fig. [155]4e).
The correlation coefficient (r) was 0.88, indicating a good predictive
power of the model. Furthermore, the metabolic profile of single-cells
was further validated in MEFs, demonstrating a high predictive power
with an AUC of 0.99 and an average accuracy of 88.42% in the
classification model (Supplementary Fig. [156]7a, b). Additionally,
there was a significant correlation between predicted and true OS
levels (r = 0.6, P < 0.0001) as shown in the correlation analysis
(Supplementary Fig. [157]7c).
Thus, through the implementation of SCLIMS technique and its
multi-modal integration, we have successfully demonstrated a link
between single-cell metabolome and cellular OS states in two different
cell types for the very first time, suggesting the potential role of
metabolome in determining cellular phenotype.
The SCLIMS unveils the causal relationship between metabolic heterogeneity
and OS status
Although there appears to be a strong correlation between the
heterogeneity of OS levels and metabolic heterogeneity, the causal
relationship between them remains unclear. We wonder if the baseline
metabolic profile determines the phenotypic or metabolic heterogeneity
of cells after they are induced to OS. We define the cells prior to OS
induction with hydrogen peroxide as “initial cells”, representing the
baseline state of both metabolism and phenotype. With SCLIMS, we
analyzed the heterogeneity of the OS levels and the single-cell
metabolome in the initial cells untreated with H[2]O[2]. We
surprisingly found that while there was minimal heterogeneity in
cellular OS levels among the initial cells (Fig. [158]5a and
Supplementary Table [159]1), there existed heterogeneity in their
single-cell metabolome (Fig. [160]5b, c). Specifically, the k-medoids
clustering analysis revealed two major metabolic subtypes (Cluster-I
and Cluster-II) in the initial cells based on their metabolomics
features (Fig. [161]5b). Through machine learning, the single-cell
metabolome can directly predict the subtype of a cell, providing
further evidence for the robust heterogeneity of metabolism in the
initial cells (Supplementary Fig. [162]8a, b). To investigate metabolic
properties of the two subtypes of initial cells, we performed a
differential analysis by comparing metabolite abundance between
Cluster-I and Cluster-II cells. A series of characteristic metabolites,
such as hypotaurine, GSH, ATP, UTP, O-phosphoethanolamine, and GSSG,
distinguishing the two subtypes were discovered (Fig. [163]5c).
Fig. 5. Metabolic heterogeneity in initial cells.
[164]Fig. 5
[165]Open in a new tab
a Distribution and box plot (inset) of DCFDA intensity in initial cells
and cells under OS. Data in distribution plot was z scored. Variance
was indicated by IQR and MAD values in Supplementary Table [166]1.
W = 58627, P < 2.2e-16 in unpaired two-tailed Wilcox rank sum test. The
data presented in the inset was normalized to the values of initial
cells. Box plots extend from 25th to 75th percentiles; central lines
represent medians; whiskers extend over 1.5 times the interquartile
range (IQR, the distance from 25th to 75th percentile); dots represent
outliers. For single-cell DCFDA intensity, a total of 1740 initial
cells (blue) and 960 oxidative stressed cells (gray) from 3 independent
experiments were analyzed. b UMAP visualization of metabolic subtypes
in initial cells. Green: cells of Cluster-I (n = 43). Purple: cells of
Cluster-II (n = 183). c Heatmap of potential metabolic markers in
Cluster-I and Cluster-II. The data was z score scaled. The color
represents relative abundance of metabolites. Yellow: relatively high
abundance; green: relatively low abundance. Each row represents a
metabolite and each column represents a cell. The representative
metabolites are labeled on the left and the clusters are labeled on the
top. ATP: adenosine triphosphate; GSH: glutathione; GSSG: oxidized
glutathione; Hypt: hypotaurine; O-PE: O-phosphoethanolamine; UTP:
uridine triphosphate. d A heatmap illustrating the metabolic similarity
between subtypes of initial cells (Cluster-I/II) and subtypes of
oxidative stressed cells (C1 to C6). Each row represents an initial
cell (subtypes are labeled on the left) and each column represents an
oxidative stressed cell (subtypes are labeled on the top). The heatmap
was plotted with similarity (1/distance) and the data was z score
scaled. The distance between cells was calculated based on single-cell
metabolome. The color represented the relative similarity: red,
relative high similarity; blue, relative low similarity. Cells of
Cluster-I is more similar to the cells with lower OS levels (cells of
C1 and part of C2). e Enrichment of GSH in cells of Cluster-I
visualized on the UMAP plot. Each dot represents a cell, the color of
the dots represents the relative GSH abundance. Red: relatively high
abundance; blue: relatively low abundance. Data was z score scaled. f
Quantification of GSH abundance in Cluster-I (n = 43, green) and
Cluster-II (n = 183, purple). Data was normalized to values in
Cluster-I. Data is represented as mean ± s.e.m. Data were collected
from at least three biological replicates. W = 7843, P = 5.85e−43 in
unpaired two-tailed Wilcox rank sum test. g The correlation of every
two metabolites were calculated (reflected as Pearson’s r) and the
network was constructed based on the correlation data for initial cells
belonging to Cluster-I (left) and Cluster-II (right) subtypes. In the
network, each node represents a metabolite while an edge connecting two
nodes indicates their correlation. The big green dot denotes GSH and
the small blue dots denote GSH correlated metabolites. Blank-colored
dots indicate metabolites without any correlation to GSH. h Rewiring
score calculated with DyNet algorithm. Metabolites with higher scores
were more rewired in topology in the correlation network of initial
cells. For (a, b, e, f), Source data are provided as Source Data files.
GSH: glutathione.
Interestingly, despite no initial cells being subjected to OS
stimulation, the metabolic disparity between the two cell subtypes in
these initial cells already demonstrated a consistent pattern with the
metabolic variation observed in cells with varying levels of OS.
Specifically, the metabolomic similarity between single cells was
determined by calculating the statistical distance of their metabolomic
data. The initial cells (from Cluster-I and Cluster-II) were paired
with each cell under OS (from C1-C6), and subsequently, the distance
was computed based on the metabolite abundance in the two cells. A
larger value of statistical distance indicates a lower degree of
similarity^[167]6. The heatmap visualization (Fig. [168]5d) was
generated based on the reciprocal value of statistical distance, which
represents the level of similarity among single cells. From a
metabolomic perspective, cells in Cluster-II exhibit greater similarity
to cells with higher levels of OS (i.e., C2-4), while cells in
Cluster-I are more similar to cells with lower levels of OS (i.e., C1)
(Fig. [169]5d). The abundance of GSH, ATP and hypotaurine were 82%,
42%, and 87% lower in cells of Cluster-II (Fig. [170]5e, f and
Supplementary Fig. [171]8c), which was in line with the above-mentioned
downregulation of GSH, ATP, and hypotaurine discovered in cells with
high OS levels (Supplementary Fig. [172]4). Metabolic pathways
including “Glutathione metabolism”, “Phosphatidylcholine Biosynthesis”,
“Phosphatidylethanolamine Biosynthesis”, “Sphingolipid Metabolism”,
“Pyrimidine Metabolism”, “Purine Metabolism”, “Thiamine Metabolism”,
“Pantothenate and CoA Biosynthesis”, and “Taurine and Hypotaurine
Metabolism” were enriched with marker metabolites in cells of Cluster-I
(Supplementary Fig. [173]8d). Notably, these pathways were all
downregulated in cells with high OS levels (i.e., cells in C6) when
compared to cells with low OS levels (i.e., cells in C1)
(Fig. [174]3e).
A remarkable demonstration of the metabolic heterogeneity exhibited by
initial cells is exemplified by GSH. The UMAP map vividly depicted the
remarkable disparity in GSH abundance among individual cells of
Cluster-I and Cluster-II (Fig. [175]5e). We further constructed
metabolic networks based by leveraging the correlation among
metabolites. The correlations between metabolites exhibited alterations
in cells belonging to Cluster-I and Cluster-II, indicating a
subtype-specific metabolic profile. For instance, the intracellular
level of GSH was strongly correlated with 15 other metabolites in
Cluster-I, whereas only 5 metabolites displayed a correlation (r > 0.8,
P < 0.05) with GSH in Cluster-II (Fig. [176]5g), indicating a notable
divergence in the GSH-centered network between the two cell subtypes.
The DyNet algorithm was employed to calculate the rewiring
score^[177]51 for each metabolite in Cluster-I and Cluster-II networks,
which quantifies modifications in a specific metabolite’s correlation
with other metabolites between the two clusters. Notably, the
identification of GSH as the most rewired metabolite (Fig. [178]5h)
highlights its pivotal role in connecting with other metabolites within
the metabolome.
The metabolic heterogeneity in initial MEFs was also investigated.
Notably, MEFs were classified into two distinct subtypes (Cluster-1 and
Cluster-2) exhibiting metabolic characteristics akin to the two
clusters observed in initial HEK293T cells (Cluster-I and Cluster-II)
(Supplementary Fig. [179]9a, b). The detailed metabolic properties of
Cluster-1 and Cluster-2 initial MEFs were further studied. Several key
metabolites, including Hypt, GSH, O-PE, and ATP exhibited enrichment in
Cluster-1 within the initial MEFs, aligning with the characteristic
metabolites in Cluster-I within the initial HEK293T cells. The Pathways
including “Glutathione metabolism”, “Phosphatidylcholine Biosynthesis”,
“Phosphatidylethanolamine Biosynthesis”, “Sphingolipid Metabolism”,
“Pyrimidine Metabolism”, “Purine Metabolism”, “Thiamine Metabolism”,
“Pantothenate and CoA Biosynthesis”, and “Taurine and Hypotaurine
Metabolism” were enriched with marker metabolites in Cluster-1
cells (Supplementary Fig. [180]10c), which was consistent with the
pathways enriched in Cluster-I in HEK293T cells (Supplementary
Fig. [181]8d). These pathways were also depleted or less enriched in
MEFs with the highest OS levels (Cluster-M6). Machine learning analysis
of the single-cell metabolome of initial MEFs confirmed the robust
heterogeneity in cellular metabolism (Supplementary Fig. [182]10a, b).
We next calculated the metabolic similarity between Cluster-1 and
Cluster-2 initial MEFs in the absence of OS with the M1-M6 MEFs under
OS conditions. Similarly, Cluster-1 initial MEFs exhibited a higher
degree of similarity to MEFs with lower levels of OS, such as M1, M2,
and part of M3 (Supplementary Fig. [183]9c). On the other hand,
Cluster-2 initial MEFs showed a greater resemblance to MEFs with higher
levels of OS including M4, M5, and M6 (Supplementary Fig. [184]9c).
These findings were consistent with the results observed in HEK293T
cells.
Next, the role of GSH in distinguishing the metabolic landscape of
Cluster-1 and Cluster-2 was confirmed in the initial MEFs. As shown in
Supplementary Fig. [185]9d, e, Cluster-1 exhibited higher levels of GSH
abundance, which could potentially serve as a discriminative factor
between the two clusters. Metabolic networks were constructed for both
Cluster-1 and Cluster-2 based on metabolite correlations (Supplementary
Fig. [186]9f). Similar to the networks observed in HEK293T cells for
Cluster-I and Cluster-II, it was found that compared to Cluster-2,
Cluster-1 displayed a more intricate network connectivity among
metabolites, indicating a subtype-specific metabolic profile within the
initial MEFs. Furthermore, the key role of GSH in governing metabolism
and correlation between metabolites, as well as its ability to
distinguish between the two subtypes of initial MEFs, were also
elucidated. These findings align with the observed role of GSH in
initial HEK293T cells. Moreover, the divergence in the GSH-centered
network between these two cell subtypes was confirmed in initial MEFs.
By employing the DyNet algorithm to compare the two metabolic networks,
GSH emerged as the fourth-highest rewired metabolite (Supplementary
Fig. [187]9g). These results underscored both the generality and
reliability of the SCLIMS technique while demonstrating its
applicability across diverse cell types.
In summary, utilizing the SCLIMS technique has revealed that while
there was no obvious heterogeneity in initial cells at the OS level,
the metabolome exhibited a high degree of heterogeneity. Interestingly,
this metabolic diversity in initial cells mirrored that observed in
cells with varying OS levels, suggesting that it may be the root cause
of heterogeneous OS levels within cells.
The metabolic heterogeneity of cells determines their senescence fate under
OS
A common fate of cellular OS is the oxidative stress-induced premature
senescence, which is a crucial type of cellular
senescence^[188]21–[189]24,[190]52. Subsequently, our objective was to
identify a live-cell probe capable of distinguishing between the cells
belonging to the two distinct metabolic subtypes, enabling us to
further investigate their respective cellular fates in OS-induced
senescence. The disparity in GSH levels between the two metabolic
subtypes (Fig. [191]5e) prompted us to explore the potential of
utilizing intracellular GSH levels as a means to distinguish between
these two cell subtypes.
The initial cells exhibiting high/low (ranging from 5% to 50%) GSH
levels were selected and mapped onto the UMAP projection alongside the
remaining initial cells, followed by calculating the proportion of
accurate matches between cells with high/low GSH levels and
Cluster-I/II (Fig. [192]6a). The fraction of correct matches exhibited
a drastic decline when the selection included more than 15% of cells
with top/bottom GSH levels (Fig. [193]6b–g), suggesting that only cells
within the range of 5%-15% accurately represented the metabolome of
Cluster-I and Cluster-II, respectively, and potentially reflected
resistance to OS and sensitivity to OS. To ensure stability, we
carefully selected cells with the highest and lowest 5% GSH levels for
further analysis, designating them as “initial cells exhibiting a
metabolome resembling that of OS-resistant cells (C1)” (MROR cells) and
“initial cells exhibiting a metabolome resembling that of OS-sensitive
cells (C6)” (MROS cells).
Fig. 6. Role of intracellular metabolic features in determining the cellular
fate in OS and OS induced senescence.
[194]Fig. 6
[195]Open in a new tab
a A flowchart showing the process of the analysis. The initial cells
were clustered based on their metabolomic features with unsupervised
clustering, revealing Cluster-I (blue palette) and Cluster-II (red
palette). Then cells with top (blue strip) and bottom (red strip)
5%-50% GSH levels were projected to the UMAP scatter plot. The correct
matches were defined as cells with top 5%-50% GSH levels to Cluster-I
(blue dots), and cells with bottom 5–50% GSH levels to Cluster-II (red
dots). Incorrect matches were labeled as green dots. Finally, the
fraction of correct matches was calculated. b–f, The visualization of
projection of cells with top/bottom 5% (b), 15% (c), 25% (d), 35% (e),
and 50% (f) GSH levels into Cluster-I and Cluster-II. Correct matches
were labeled as blue (cells with top 5–50% GSH levels vs Cluster-I) and
red (cells with bottom 5%-50% GSH levels vs Cluster-II) dots. Incorrect
matches were labeled as green dots. Cells with intermediate GSH levels
were labeled as gray dots. g Quantification of fraction of accurate
matches between cells with top/bottom 5%-50% GSH levels and Cluster-I
and Cluster-II. Blue: correct matches; green: incorrect matches. h,
Experimental setup of FACS separation of MROR and MROS cells and gating
of the FACS: cells with top 5% (MROR, blue) and bottom 5% (MROS, red)
GSH intensity were collected according to their fluorescent intensity.
i, j Representative images (i) and quantification (j) of DCFDA staining
of MROR and MROS cells before and after OS induction. F(1, 8) = 7.405,
P = 0.0262 in two-way ANOVA. k, l Representative images (k) and
quantification (l) of SA-β-Gal staining of MROR and MROS cells before
and after induction of OS-induced senescence. F(1, 8) = 9.177,
P = 0.0163 in two-way ANOVA. Scale bar, 50 μm. P values in two-way
ANOVA with Turkey’s multiple comparisons were labeled in the plot.
n = 3 for each group. All P values were reported as multiplicity
adjusted P values for multiple comparisons. Data was normalized to the
values of MROR group in control cells. Data is presented as mean ±
s.e.m. For (j, l), Source data are provided as Source Data files. Blue:
MROR cells; red: MROS cells. OS: oxidative stress. MROR cells: initial
cells exhibiting a metabolome resembling that of OS-resistant cells.
MROS cells: initial cells exhibiting a metabolome resembling that of
OS-sensitive cells.
To isolate the MROR and MROS cells, we employed a live-cell GSH
fluorescent dye (monochlorobimane, mClB)^[196]53 and fluorescence
activated cell sorting (FACS). The MROR cells (Top 5% fluorescent
intensity) and the MROS cells (Bottom 5% fluorescent intensity) were
collected by the FACS according to their mClB fluorescent intensity
(Fig. [197]6h and Supplementary Figs. [198]11, [199]12). The DCFDA
intensity (Fig. [200]6i, j) had only 3% difference between MROR and
MROS cells. However, following OS induction, the MROS cells exhibited
2.7-fold higher of DCFDA intensity compared to their MROR counterparts
(Fig. [201]6i, j), indicating an elevated state of OS. Furthermore,
SA-β-Gal staining confirmed the sensitivity of MROS cells in OS-induced
senescence (Fig. [202]6k, l). Thus, the data from the SCLIMS suggest
that the metabolic features of an initial cell may dictate its destiny
under OS and OS-induced senescence.
The key metabolites identified by SCLIMS mitigate OS and cellular senescence
The aforementioned results suggest that the downregulation of critical
metabolites, identified by the SCLIMS, may be a contributor to OS,
indicating the potential for reversing OS and OS-induced senescence
through targeted metabolite supplementation. To explore this
possibility, we opted to conduct an experiment using three key
metabolites: hypotaurine, phosphocreatine, and O-phosphoethanolamine,
which all showed similar characteristics such as declining in OS
(Fig. [203]2b), distributing heterogeneously across metabolic subtypes
(C1-6) (Fig. [204]3d), and serving as metabolic markers distinguishing
the initial cells with different fates (Fig. [205]5c).
Treatment of cells with the three metabolites resulted in an average of
67% reduction in OS levels, as evidenced by a decrease in DCFDA
intensity within the cells (Fig. [206]7a, b). OS is a common inducer of
senescence^[207]22,[208]54,[209]55 and also plays a vital role in
natural aging^[210]56. We therefore explored the effects of these key
metabolites on OS-induced senescence. The SA-β-Gal staining showed that
hypotaurine, phosphocreatine and O-phosphoethanolamine considerably
decreased the cellular senescence (Fig. [211]7c, d and Supplementary
Fig. [212]13a–c). The staining intensity exhibited an average of 48%
reduction in key metabolite-treated cells compared with vehicle-treated
OS cells. The growth arrest of cells, a marker for cellular
senescence^[213]55,[214]57, was also rescued by these metabolites
(Fig. [215]7e, f and Supplementary Fig. [216]13d–f). The mitochondrial
membrane potential (MMP), which is compromised in cellular
OS^[217]58,[218]59, was recovered by targeted supplement of key
metabolites (Fig. [219]7g, h). Furthermore, the examination of the
three key metabolites on MEFs under OS yielded analogous outcomes,
thereby indicating the generalized applicability of these key
metabolites across diverse cell types (Supplementary Fig. [220]14).
Collectively, these findings imply that the SCLIMS-identified
metabolites exert a protective effect by reducing the OS level and
alleviating senescence.
Fig. 7. Effects of key metabolites on OS and induced senescence.
[221]Fig. 7
[222]Open in a new tab
a, b Representative images (a) and quantification (b) of DCFDA staining
of control cells and oxidative stressed cells with indicated
treatments. n = 5 for each group. Scale bar, 50 μm. F(4, 20) = 5.991,
P = 0.0024 in One-way ANOVA. P values in one-way ANOVA with multiple
comparison were labeled in the plot. Data was normalized to values of
control group. c, d Representative images (c) and quantification (d) of
SA-β-Gal staining of control and oxidative stressed cells with
indicated treatments. n = 9, 9, 3, 3, and 3 for Control, Vehicle
treated, Hypt treated, PCr treated, and O-PE treated group
respectively. Scale bar, 50 μm. F(4, 22) = 17.29, P = 5.50e-9 in
One-way ANOVA. P values in one-way ANOVA with multiple comparison were
labeled in the plot. Data were normalized to values of control group.
e, f Representative images (e) and growth curve (f) of control and
oxidative stressed cells with indicated treatments at indicated time
points. Growth curve was plotted with at least 15 random fields from 3
independent biological replicates for each group at each indicated time
point. Scale bar, 50 μm. F(4, 520) = 41.26, P < 2.2e−16 in two-way
ANOVA. P values in two-way ANOVA with Turkey’s HSD comparison were
labeled in the plot. Each group was compared with vehicle-treated
oxidative stressed cells. Data were normalized to values of control
group at 0 h. g, h Representative images (g) and quantification (h) of
TMRE staining of control and oxidative stressed cells with indicated
treatments. n = 9, 9, 5, 5, and 4 for Control, Vehicle treated, Hypt
treated, PCr treated, and O-PE treated group respectively. Scale bar,
50 μm. F(4, 27) = 26.12, P = 6.14e−9 in one-way ANOVA. P values in
one-way ANOVA with multiple comparison were labeled in the plot. Data
was normalized to values of control group. All data are presented as
mean ± s.e.m. OS: oxidative stressed cells; Hypt: hypotaurine (1 mM);
PCr: phosphocreatine (0.5 mM); O-PE: O-phosphoethanolamine (40 μM). All
P values were reported as multiplicity adjusted P values for multiple
comparisons. For (b, d, f, h), Source data are provided as Source Data
files. Blue: control; red: OS+vehicle; green: OS+Hypt; purple: OS+PCr;
yellow: OS + O-PE.
Treatment of key metabolites regulated the metabolome of single cells
To investigate the precise alterations in the metabolome of individual
cells upon treatment with key metabolites, we conducted metabolic
analysis on cells from five groups: Non-OS, vehicle-treated OS,
Hypt-treated OS, PCr-treated OS, and O-PE-treated OS. Subsequently, we
examined the distinct metabolic characteristics exhibited by these cell
populations (Supplementary Fig. [223]15a). Notably, cells treated with
key metabolites were positioned between non-OS and vehicle-treated OS
cells, indicating an intermediary metabolic state between the two
groups. In consistency, cells treated with key metabolites exhibited
intermediate intensity in DCFDA and SA-β-Gal staining along with
intermediate growth rate when compared to non-OS and OS cells
(Fig. [224]7a–f). This suggests that the treatment of metabolites may
have partially restored the disrupted metabolism in oxidative stressed
cells and further alleviated cellular oxidative stress and senescence.
To quantify the similarity between the metabolome of metabolite-treated
cells and non-OS/OS cells, we calculated the paired distance between
these cell types. Interestingly, compared to non-OS and OS cells, the
cells treated with key metabolites exhibited a smaller distance to
non-OS cells (Supplementary Fig. [225]15b). These findings indicate
that treatment with key metabolites modulated the metabolome of OS
cells towards a state similar to that of non-OS cells.
Next, the detailed alteration of metabolome in OS and
metabolite-treated cells was further studied. By conducting a
comparative analysis of metabolites in non-OS cells and those treated
with OS, we identified a series of downregulated metabolites under OS
conditions, which were subsequently restored upon treatment with key
metabolites (Supplementary Fig. [226]15c). For instance, some key
metabolites downregulated in OS, such as hypotaurine, phosphocreatine,
O-phosphoethanolamine, GSH, ATP (Figs. [227]2b, [228]3d), were
recovered under the treatment of hypotaurine, phosphocreatine, and
O-phosphoethanolamine (Supplementary Fig. [229]15c). The metabolic
pathways involved in the recovery of the metabolome were dissected
through MSEA analysis using the recovered metabolites under treatment
(Supplementary Fig. [230]15d). A subset of these metabolic pathways
overlapped with those downregulated in OS. Specifically, the “Citric
acid cycle” in mitochondrial metabolism; “Phosphatidylethanolamine
Biosynthesis,” “Phosphatidylcholine Biosynthesis,” and “Sphingolipid
Metabolism” in lipid metabolism; “Aspartate metabolism” and “Glutamate
metabolism” in amino acid metabolism; “Pyrimidine metabolism” and
“Purine metabolism” in nucleotide metabolism; and “Amino sugar
metabolism,” “Galactose metabolism,” and “Nucleotide sugars metabolism”
in sugar and derivatives metabolism were all enriched with recovered
metabolites and overlapped with pathways downregulated in OS.
Consistently, OS cells treated with key metabolites exhibited lower
levels of OS, reduced SA-β-Gal intensity, and improved mitochondrial
function (Fig. [231]7 and Supplementary Fig. [232]14).
The collective findings suggest that the modulation of crucial
metabolites governs the cellular metabolome under OS, leading to the
restoration of a multitude of metabolites. Treatment with these pivotal
metabolites induces a metabolic state resembling that of non-OS cells,
thereby highlighting their significant role in regulating OS and
senescence pathways. Consequently, it can be inferred that specific key
metabolites exert control over the cellular metabolome. The observed
extensive heterogeneity and metabolic disparities in OS cells may stem
from variations in the abundance levels of these essential metabolites.
The SCLIMS-identified protective metabolites promote healthy aging and extend
lifespan
The effect of key metabolites treatment across two different cell types
confirmed the regulatory role of metabolism in OS and senescence,
leading to the question whether key metabolites further regulate animal
aging. To explore the regulatory effects of metabolism on animals, we
introduced the C. elegans aging model^[233]60,[234]61 to investigate
the impact of the key metabolites (hypotaurine, phosphocreatine, and
O-phosphoethanolamine) on the process of natural aging (Fig. [235]8a).
A serial of concentrations of the three metabolites were added into the
Nematode Growth Medium (NGM) at the age of L4 and throughout the
lifespan of the worms. The OS level, lifespan and healthspan of the
worms were then evaluated. Strikingly, the supplement of hypotaurine,
phosphocreatine and O-phosphoethanolamine at various doses caused a
remarkable extension in the lifespan of C. elegans by approximately
33%-50% (Fig. [236]8b and Supplementary Fig. [237]16a).
Fig. 8. Metabolic intervention extends lifespan and promotes healthy aging in
C. elegans.
[238]Fig. 8
[239]Open in a new tab
a A flowchart of experimental setup of lifespan and healthspan assay of
C. elegans. b Lifespan of C. elegans treated with vehicle (n = 221),
0.4 mM hypotaurine (Hypt) (n = 320), 0.2 mM phosphocreatine (PCr)
(n = 225) and 0.1 mM O-Phosphoethanolamine (O-PE) (n = 387). P values
in two-tailed log rank test compared with vehicle-treated worms were
labeled in the plot, P < 2.2e−16 for all comparisons indicated in (b).
c, d Representative images (c) and quantification (d) of DHE staining
of L4 (young adult) (n = 14) and Day-9 (aged) worms with indicated
treatments (n = 21, 23, 19, and 24 for vehicle, Hypt, PCr, and O-PE
treated worms respectively). Scale bar, 200 μm. F(4, 96) = 65.74,
P < 2.2e-16 in One-way ANOVA. P values in One-way ANOVA with multiple
comparison were labeled in the plot. P values were reported as
multiplicity adjusted P values for multiple comparisons. Data was
normalized to values of vehicle treated group at Day 9. Data is
presented as mean ± s.e.m. e, f Representative images (e) and
quantification (f) of L4 (n = 12) and aged C. elegans thrashing under
treatment of vehicle (n = 19), 0.4 mM hypotaurine (Hypt) (n = 28),
0.2 mM phosphocreatine (PCr) (n = 23) and 0.1 mM O-Phosphoethanolamine
(O-PE) (n = 13). Arrows indicate immobilized worms. Scale bar, 1 mm.
F(4,90) = 27.58, P = 6.00e−15 in One-way ANOVA. P values in One-way
ANOVA with multiple comparison were labeled in the plot. P values were
reported as multiplicity adjusted P values for multiple comparisons. g,
h Representative traces (g) and quantification (h) of free moving C.
elegans. The traces showed the track of free moving worms in 1 min. At
day 1, data of tracks was derived from 294, 368, 317, and 391 worms for
vehicle, Hypt treated, PCr treated, and O-PE treated group,
respectively. At day 5, data of tracks was derived from 184, 284, 175,
and 305 worms for vehicle, Hypt treated, PCr treated, and O-PE treated
group, respectively. At day 9, data of tracks was derived from 176,
309, 316, and 153 worms for vehicle, Hypt treated, PCr treated, and
O-PE treated group, respectively. Scale bar, 1 mm. F(3,26296) = 73.15,
P = 4.17e−47 in Two-way ANOVA. P values in Two-way ANOVA with Turkey’s
HSD comparison (vehicle vs O-PE, Hypt, and PCr, respectively) were
labeled in the plot. P values were reported as multiplicity adjusted P
values for multiple comparisons. Data were collected from 3 independent
biological replicates. For (b, d, f, h), Source data are provided as
Source Data files. For (b and h), blue: Vehicle treated worms; green:
Hypt treated worms; purple: PCr treated worms; yellow: O-PE treated
worms. For d and f, gray: L4 worms; red: vehicle treated aged worms;
green: Hypt treated aged worms; purple: PCr treated worms; yellow: O-PE
treated aged worms. Hypt: 0.4 mM hypotaurine; PCr: 0.2 mM
phosphocreatine; O-PE: 0.1 mM O-Phosphoethanolamine.
The OS level of the aged C. elegans were assessed by dihydroethidium
(DHE) staining^[240]62. The findings unveiled a remarkable elevation in
OS levels among aged worms, as indicated by the intensity of DHE.
However, this surge in OS was mitigated upon supplementation with the
three metabolites in C. elegans (Fig. [241]8c, d). The intensity of DHE
was reduced by an average of 16% in metabolite treated worms.
It has been reported that the mobility of worms is drastically
compromised in aging as a sign of deterioration of health^[242]63. We
next examined the locomotion of worms treated with and without these
metabolites from the L4 stage onwards. Aged nematodes (Day 9) were
subjected to the “thrashing” assay, wherein their swimming movements
were observed to assess their physical mobility^[243]63. The elderly
worms exhibited a 43% decline in their thrashing rate, which was
effectively ameliorated by the administration of hypotaurine,
phosphocreatine and O-phosphoethanolamine (Fig. [244]8e, f and
Supplementary Fig. [245]16b).
Another indication of the deterioration in health of aged C. elegans is
the reduction in their speed of movement^[246]64. The worms at three
different ages were analyzed and a progressively decline in free moving
speed was indeed observed (Fig. [247]8g, h). The supplementation of the
three metabolites from L4 improved the free moving speed of the aged
worms (Fig. [248]8h and Supplementary Fig. [249]16c, d). Free moving
speed of metabolite-treated worms was 1.4–1.8 fold higher compared with
vehicle-treated worms at Day 9, and 1.1–1.2 fold higher at Day 5.
Thus, the key metabolites identified by SCLIMS, including hypotaurine,
phosphocreatine, and O-phosphoethanolamine, possess remarkable
potential to prolong lifespan and foster graceful aging in C. elegans
by effectively preventing cellular OS and senescence.
Discussion
Emerging evidence indicates a diversity of cellular subtypes in
senescence^[250]7,[251]8,[252]65,[253]66, cancer^[254]6,[255]67,
diabetes^[256]68,[257]69, and inflammatory diseases^[258]70, suggesting
that there is vast heterogeneity among cells in various biological
processes. However, previous studies on cellular metabolism have
predominantly been performed at the homogenate level, potentially
disregarding the metabolic heterogeneity and intricate metabolic
changes of individual cells. Due to the challenges in obtaining
metabolomic information from individual live cells, determining
metabolic heterogeneity in single cells has proven to be a formidable
task. The SCMS technique we have previously established excels at
unraveling the metabolome at a single-cell resolution^[259]17,[260]18.
However, it encounters challenges when it comes to correlating the
single-cell metabolome with cellular function and phenotype. Recently,
the integrative analysis across multiple modalities brought new
insights into cellular heterogeneity and elucidated the underlying
mechanisms governing biological processes^[261]11,[262]71–[263]74,
thereby illustrating the crucial role of cross-modality analysis. In
this study, we established an approach called SCLIMS by combining SCMS
and live-cell imaging to the metabolome of individual cells with their
cellular OS status, thereby enabling a cross-modality analysis of both
metabolomics profiles and cellular phenotypes at single-cell
resolution. This technology provides some notable contributions.
Firstly, with SCLIMS we unveiled distinct metabolic signatures among
the six subtypes of cells under OS, each with specific oxidative
levels. The detailed single-cell metabolic profile of OS has been
dissected, revealing the deterioration of metabolic processes
associated with redox balance, energy metabolism, lipid metabolism and
mitochondrial function. The SCLIMS has not only confirmed various
alterations in metabolism under OS as reported in previous studies, but
also unveiled discoveries of metabolic changes, including modifications
in amino acid metabolism and the transition from the “Malate-Aspartate
Shuttle” to the more sophisticated “Glycerol Phosphate Shuttle”.
Secondly, the utilization of machine learning analysis on the
single-cell metabolome substantiates the predictive capacity of
individual metabolic characteristics regarding cellular heterogeneity
and phenotype. Thirdly, the SCLIMS has led to the discovery that the
ultimate destiny of cells following OS induction is determined by their
initial heterogeneity in metabolomics. Lastly, the key metabolites
identified by SCLIMS exhibit protective effects against OS, cellular
senescence, and natural aging. Overall, the SCLIMS technique sheds
lights into the study of metabolic changes in OS and therapeutic
interventions in aging.
In the present study, the SCLIMS shows that the heterogenous states of
cells can be predicted directly with their intracellular metabolome,
showing a tight link between metabolic features and cellular phenotype.
In addition, a more remarkable discovery revealed by the SCLIMS is that
the destiny of cells under OS can be determined by their initial
metabolomics status. This technology has verified that GSH is strongly
correlated with many metabolites in the metabolome and may play a key
role in cell fate determination, as evidenced by the fact that GSH-rich
cells exhibit greater resistance to OS. However, it should be noted
that such resistance is not solely attributed to GSH; rather, our
SCLIMS analysis reveals elevated levels of other key metabolites such
as hypotaurine, and O-phosphoethanolamine in cells exhibiting enhanced
oxidative resistance. Indeed, the anti-oxidant effect of these
metabolites has been further confirmed in the aged C. elegans. In
essence, the GSH-cored metabolome serves as the determinant of cell
phenotype and fate in OS. Among the top 15 rewired metabolites, GSH is
well correlated with the other 13 metabolites including glutamate,
creatine, glutamine, taurine, threonine, N-acetyl-aspartic acid,
aspartic acid, asparagine, UDP-N-acetylglucosamine, proline, GABA,
cystathionine, and glucose. These metabolites participate amino acids
metabolism as well as carbohydrate metabolism, which were compromised
in cells with higher OS levels (Fig. [264]3e). Other studies reported
the effects of these top-rewired metabolites in aging and senescence as
well. For example, taurine was reported to decline in aging and
supplement of taurine increases healthspan and lifespan in various
species^[265]75. Taurine protected telomerase and mitochondrial
function, along with decreasing inflammation and DNA damage. Creatine
was reported to promote healthy aging by attenuating inflammation and
preventing bone mineral loss^[266]76. Creatine improved neuronal
function by antioxidant effect and exhibited therapeutic effect against
age-related diseases including Alzheimer’s disease, Parkinson’s
disease, and heart failure^[267]77. The level of threonine, aspartic
acid, and proline were reported to be positively correlated with
lifespan of yeasts^[268]78. Glutamine promotes autophagy via AMPKα
lactylation and suppresses senescence^[269]79. N-acetyl-aspartic acid
was reported to decline in aging and was related to brain
atrophy^[270]80. Asparagine prevented stem cell aging by regulating the
autophagy-lysosome pathway^[271]81. The underlying mechanism behind the
notable disparity in metabolic characteristics among initial cells
remains elusive and necessitates further investigations. However, a
possible explanation is that it could be a result of the asymmetric
division of cytoplasm during cell division^[272]82–[273]85. Another
possibility may be attributed to differences in cellular contact,
micro-environmental development, or potential transport of certain
metabolites between neighboring cells that has gone undetected^[274]86.
However, from a metabolic view, abundance of key metabolites determined
the metabolomic profile of cells and recovered the disrupted metabolic
pathways and the metabolome under OS along with reducing the OS and
senescent level (Fig. [275]7 and Supplementary Figs. [276]14, [277]15),
suggesting the role of key metabolites in determining the metabolic and
phenotypic heterogeneity.
The SCLIMS-identified key metabolites provides opportunities in
senescence and aging intervention. The OS-induced senescence is one of
the important models of cellular senescence^[278]22,[279]54,[280]55 and
play crucial role in various diseases^[281]87,[282]88. The
supplementation of the key metabolites, including hypotaurine,
phosphocreatine, and O-phosphoethanolamine, can effectively mitigate
the cellular OS and the OS-induced senescence. Moreover, these
metabolites also prevent OS, prolong lifespan, promote healthy aging
and delay the decline in mobility during aging in C. elegans. Combining
the clues from the literature, we posit that these metabolites may
regulate cellular OS and senescence through multiple mechanisms. For
instance, hypotaurine serves as a hydrogen donor for NAD^+ during its
conversion into taurine, thereby generating NADH as a
by-product^[283]31. This process effectively restores redox balance in
the presence of OS. Phosphocreatine serves as a direct catalyst for the
conversion of ADP into ATP^[284]89, functioning as a quintessential
cellular energy reservoir that possesses the inherent capability to
reinstate equilibrium in energy levels. O-phosphoethanolamine has been
reported to mitigate mitochondrial dysfunction^[285]90 and effectively
restore membrane, as it serves as the fundamental precursor of membrane
lipids^[286]91. Therefore, these metabolites may potentially impede
senescence by modulating various cellular processes, including energy
metabolism, mitochondrial function, and lipid metabolism. The
deficiency of such metabolites may render cells more susceptible to
senescence-inducing factors, such as OS. Furthermore, a more paramount
consequence of the metabolic intervention lies in its ability to
prolong healthspan, which assumes a relatively pivotal role in the
realm of aging research when compared to lifespan^[287]63,[288]92. The
decline of physical function is a common occurrence in both the early
and late stages of aging, and bestowing longevity upon frailty offers
minimal advantage to the individual^[289]93. Therefore, screening for
potential metabolites from our SCLIMS database presents an opportunity
for promoting health benefits during the aging process.
The current study has certain limitations that we would like to
address, along with potential solutions for future studies. Firstly,
the identification of metabolites at the single-cell level poses a
significant challenge due to the complexity of MS/MS analysis.
Acquiring MS/MS spectra for hundreds of m/z in the metabolome is indeed
arduous. However, there are promising avenues to enhance metabolite
identification efficacy. For instance, ion mobility mass spectrometry
can potentially differentiate metabolites sharing identical m/z values
by considering collision cross section^[290]94–[291]96. Additionally,
optimizing scan speed and extending sampling duration can facilitate
acquiring comprehensive MS/MS spectra from single cells^[292]97.
Secondly, the SCLIMS utilized in this current study was meticulously
designed to incorporate cultured cells and cellular models of OS. The
versatility of SCLIMS extends to tissue-embedded cells, as they can
also be effectively labeled with fluorescence markers. However, the
potential application of SCLIMS in tissue-embedded cells remains an
intriguing area for future exploration and investigation. Thirdly, one
must acknowledge the challenges associated with analyzing fixed cells
when employing the SCLIMS technique. Nevertheless, by enabling analysis
of fixed cells, a myriad of phenotypic features such as SA-β-Gal
staining, immunofluorescence analysis, and immunohistochemistry can be
seamlessly integrated into the cell metabolome profiling process.
In summary, this study presents a cross-modality analysis integrating
single-cell metabolomic profile and cellular phenotype enhancing our
understanding of cell heterogeneity and subtype-specific metabolic
signatures in a cellular model of OS. Most importantly, the
cross-modality platform and analysis described in this study provide a
way in single-cell research. The single-cell metabolome and the
cellular phenotype such as OS status are directly linked and
integrated. The heterogeneous states are explained with single-cell
metabolome and metabolic pathways. Significantly, the insights into the
metabolic regulation governing OS, cellular senescence, and natural
aging serves as a valuable resource for future investigations into
interventions targeting oxidative damage, aging and senescence.
Furthermore, this cutting-edge platform possesses the remarkable
capability to integrate single-cell metabolomics profiling with a
diverse array of cellular phenotypes assessed by live-cell labeling.
For instance, (1) The SCLIMS can be combined with live-cell
mitochondrial probes such as probes for mitochondrial membrane
potential (i.e. TMRE probe) and mitochondrial morphology (i.e.
Mito-Tracker). This enables the study of the interaction between
mitochondrial function and the metabolome at single-cell level; (2) The
SCLIMS can be integrated with calcium imaging^[293]98 which labels
neuronal activities and enables the performance of multi-modal analysis
of heterogeneity in metabolome and neuronal functions; (3) The SCLIMS
can be utilized to investigate the correlation between cellular
metabolome and cell cycle by incorporating dynamic live-cell
fluorescent probes for real-time monitoring of cell division and
proliferation^[294]99. Thus, with any technique labeling live cells
with fluorescent, this cross-modality platform will become a feasible
way for integrative analysis and a powerful tool for the discovery of
secrets in single cells.
Methods
Chemicals
NaCl, KCl, CaCl[2], MgCl[2], HEPES, NaOH, sucrose, NH[4]HCO[3],
Na[2]HPO[4], KH[2]PO[4], K[2]HPO[4], MgSO[4], cholesterol, hypotaurine,
and O-phosphoethanolamine were purchased from Sigma-Aldrich. Phosphate
buffer saline was purchased from Sangon Biotech. Dulbecco’s Modified
Eagle’s medium (DMEM) was purchased from HyClone. Fetal bovine serum
and trypsin-EDTA (0.25%) were purchased from Gibco. Trypan blue,
penicillin and streptomycin were purchased from Biosharp. Hydrogen
peroxide was purchased from Sinopharm. Phosphocreatine was purchased
from Aladdin. The Senescence-Associated β-Galactosidase kit and
Mitochondrial membrane potential assay kit were purchased from
Beyotime. A cellular ROS assay kit and dihydroethidium (DHE) were
purchased from Abcam. Live-cell GSH probe (mClB) was purchased from
MedChemExpress.
Cell culture
HEK293T cell line were originally obtained from ATCC (CRL-3216). The
cell line was authenticated by ATCC with STR profiling. Primary MEFs
were a kind gift from Professor Chunlei Cang in University of Science
and Technology of China. All cells were cultured in Dulbecco’s Modified
Eagle’s medium (DMEM) (HyClone), supplemented with 10% fetal bovine
serum (FBS, Gibco) and 100 U/ml penicillin 100 µg/ml streptomycin
(Biosharp) at a temperature of 37 °C, with 5% CO[2] in a humidified
atmosphere. The culture medium was refreshed every 2-3 days, and the
cells were subcultured every 3-5 days when they reached approximately
80% confluency.
C. elegans strain and maintenance
The Caenorhabditis elegans (C. elegans) were cultured and maintained on
Nematode Growth Medium (NGM) seeded with E. Coli OP50 at 20 °C. N2
(wild isolate) strain was used in all C. elegans experiments. Plates
were maintained by transferring the worms every 3 days. For DHE
staining and thrashing analysis, worms at L4 and at Day 9 after L4 were
used. For lifespan analysis, the survival of worms was observed
throughout the whole lifespan. For activity analysis, worms at Day 1,
Day 5 and Day 9 after L4 were used.
Oxidative stress model
Cells were seeded and allowed to grow overnight. Subsequently, they
were treated with hydrogen peroxide at a final concentration of 80 μM
for HEK293T cells and 240 μM for MEFs in the culture medium for 1 h,
followed by replacement with fresh medium. Finally, the cells were
cultured for an additional 48 h to establish an oxidative stress model.
Treatment of cells with metabolites
Cells were seeded and allowed to grow overnight. Subsequently, the
cells were treated with hydrogen peroxide at a final concentration of
80 μM for HEK293T cells and 240 μM for MEFs in culture medium for 1 h.
After that, the medium was replaced with fresh medium supplemented with
specific metabolites at indicated concentrations. The cells were then
cultured for an additional 48 h before further assays.
Metabolite treatment and life span assay of C. elegans
Life span assays of C. elegans were conducted at a temperature of
20 °C. Metabolite treatment was administered by adding specific
metabolites at the indicated concentrations to NGM plates, which were
then incubated overnight prior to use. Following bleaching,
age-synchronized eggs were washed with M9 buffer and subsequently
placed on NGM plates. Late L4 larvae or young adult worms were
subsequently transferred to NGM plates that had been seeded with
heat-inactivated OP50 E. coli and supplemented with 0.1 mg/ml of
5’-FUDR, as well as the indicated treatment of metabolites.
Approximately 100 worms were placed on each plate, which was then
inspected and scored every one to two days. The worms were moved to
fresh plates every one to two days in order to ensure the efficacy of
the drugs and metabolites. Worms that exhibited no response to
mechanical stimulation were considered deceased. Worms displaying a
“protruding vulva”, those that were lost, or had burrowed into the
medium were censored. Statistical analysis was conducted using the
MATLAB function “logrank”
([295]www.mathworks.com/matlabcentral/fileexchange/22317), and P values
were calculated.
Behavioral analysis of C. elegans
The ‘thrashing’ assay was employed to assess the locomotion of C.
elegans. On Day 9 post-adulthood, worms subjected to specific
treatments were transferred to M9 buffer and allowed to acclimate for
1 min before body bends were quantified using a dissecting microscope.
The physical function of C. elegans was assessed by monitoring the
locomotion of the worms on NGM plates. On Day 1, Day 5, and Day 9
post-adulthood, freely moving worms subjected to specific treatments
were recorded using a digital camera and analyzed in ImageJ (version
1.54 g, [296]https://imagej.nih.gov/ij/) with the ‘wrMTrck’ plugin as
per manual instructions. The traveling speed was then calculated using
the plugin.
Cell viability assay
Cell viability assay was performed according to the manufacturer’s
manual. Cells were dissociated using trypsin-EDTA (0.25%) (Gibco) for
1 min at 37 °C, followed by termination of the dissociation process
through the addition of an equal volume of DMEM supplemented with 10%
fetal bovine serum. Cells were suspended, centrifuged, and then
resuspended with PBS. Subsequently, they were stained with trypan blue
(Biosharp) at a final concentration of 0.04%. The cells were then
enumerated under a microscope; the stained cells were designated as
non-viable. Cell viability was determined by calculating the ratio of
unstained cells to the total number of both stained and unstained
cells.
SA-β-Gal assay
The experimental procedure was performed in accordance with the
protocols provided by the SA-β-Gal assay kit (Beyotime, China).
Briefly, cells were washed with phosphate-buffered saline (PBS) and
fixed with fixing reagents at room temperature for 15 min. After three
washes with PBS for 3 min each, staining solution was prepared
according to the manufacturer’s instructions prior to use. Cells were
stained overnight at 37 °C, and images of five randomly selected fields
were captured using a bright field setting for subsequent analysis.
Oxidative stress assay
To evaluate the extent of cellular oxidative stress, a live-cell probe
DCFDA was prepared according to the manufacturer’s protocol (Abcam).
Subsequently, live cells were incubated with a 10 μM DCFDA solution for
25 min at 37 °C and 5% CO[2], followed by PBS washing. Images were
promptly captured using a fluorescent microscope (Leica), and five
random fields were selected for analysis in each dish.
To assess the level of oxidative stress in C. elegans, worms were
exposed to a final concentration of 3 μM DHE (Dihydroethidium) in M9
buffer at 20 °C for 30 min, followed by washing with M9 buffer.
Subsequently, the worms were transferred onto glass slides and imaged
using a fluorescent microscope (Leica).
Mitochondrial membrane potential assay
TMRE was utilized to assess the mitochondrial membrane potential (MMP)
of viable cells in accordance with the manufacturer’s instructions.
Briefly, cells were incubated with 1X TMRE (Beyotime) in serum-free
DMEM for 15 min at 37 °C. Subsequently, the cells were rinsed with warm
serum-free DMEM and immediately imaged using a fluorescent microscope
(Leica, version 4.6.2 build: 410). Images of five randomly selected
fields were captured for analysis.
Analysis of SA-β-Gal intensity
The SA-β-Gal-stained area in images of random fields was extracted
using the ‘IHC toolbox’ plugin in ImageJ, resulting in a new image of
the stained area. The color images of the extracted SA-β-Gal stained
area were then converted to 8-bit grayscale and calibrated within
ImageJ (version 1.54g). Finally, the gray value (integrated density) of
the stained area was calculated using ImageJ. The intensity of SA-β-Gal
was quantified by calculating the gray value (integrated density) of
the stained area normalized to the total cell area in each image.
Analysis of fluorescence intensity
For random field images, color images of cells stained with fluorescent
live-cell probes (DCFDA and TMRE) were imported into ImageJ software.
The images were then converted to 8-bit grayscale and calibrated before
being thresholded. The fluorescence intensity was calculated as the
mean gray value of cells in the field by measuring the integrated
density of fluorescent positive area divided by the fluorescent
positive area. For single cells, the image was converted to an 8-bit
grayscale image and calibrated. Fluorescent intensity was analyzed
using ROI manager in ImageJ, which allowed for analysis of individual
cells through selection. The mean gray value for each cell was
calculated by dividing the integrated density of a cell by its area.
For C. elegans, the level of oxidative stress (indicated by DHE
fluorescence) was quantified using ImageJ software. The color images
were converted to 8-bit grayscale and calibrated prior to thresholding.
The gray value (integrated density) of fluorescent positive areas in
individual worms was then calculated and normalized by the
corresponding area.
The workflow and experimental setting of SCLIMS
The SCLIMS platform comprises two primary components: live-cell imaging
and single-cell MS. Cells were initially stained with a 10 μM DCFDA
solution for 25 min at 37 °C and 5% CO[2], followed by PBS washing.
Subsequently, the cells were captured using a fluorescent microscope to
record their spatial distribution. The acquired images were
subsequently subjected to analysis, enabling the calculation of
oxidative stress levels in individual cells.
The cells were then transferred to the single-cell patch clamp platform
and incubated in a bath solution containing 140 mM NaCl, 5 mM KCl, 2 mM
CaCl[2], 1 mM MgCl[2] and 10 mM HEPES (pH adjusted to 7.4 with NaOH;
∼320 mosmol with sucrose) and approached by a borosilicate glass
pipette filled with pipette solution (88 mM NH[4]HCO[3]) using a
micromanipulator. The cells were selected based on the fluorescent
images acquired in the preceding step. The cells were patched with a
high-quality seal (>1 GΩ) and the cell membrane was disrupted by rapid
application of negative pressure. Mild negative pressure was then
applied to the pipette to obtain cytoplasmic chemical constituents,
which were subsequently analyzed using mass spectrometry after removal
of the pipette from the bath solution.
Following the extraction of cellular cytoplasmic constituents, the
capillary was connected to a MS system as described below. An AC
voltage of 4 kV amplitude and approximately 500 Hz frequency was
applied externally to the spray capillary micropipette, while
maintaining a distance of approximately 5 mm between the tip of the
spray micropipette and the orifice of the MS instrument.
High-resolution mass measurements were analyzed using a Q Exactive Plus
MS instrument (Thermo Fisher Scientific, San Jose, CA, USA). nanoESI
source and Orbitrap mass analyzer were used. The main experimental
parameters were established as follows: capillary temperature at
275 °C, S-lens radio frequency (RF) level set to 50%, mass resolution
of 70,000, maximum injection time of 10 ms, AGC target of 1e6, and
microscan rate of 1. Negative ion mode was employed throughout all
experiments. Data was acquired under full scan mode. For MS/MS,
collision energy was set to hcd = 30. Data was collected with Thermo
Scientific Exactive Tune software (version 2.9.0.2926). The MS data
were then processed. Data of each single cell was paired with oxidative
stress levels based on the fluorescent images.
Single-cell mass spectrometry data processing
The spectral data of individual cells were initially stored in separate
files by the instrument. Subsequently, all files were converted into
mzML format using ProteoWizard (version 3.0.9870). The XCMS package
(version version 3.9.1) in R statistical environment was utilized to
process all single-cell MS data, encompassing peak calling, peak
alignment, and quality control. A signal-to-noise (S/N) filter of 3 was
applied to the m/z signal, and metabolic signals were identified as
those with a frequency exceeding 20% across all tested cells. The data
was organized into a matrix, where metabolites were represented by rows
and cells were represented by columns. All intensities were normalized
to the total ion current (TIC) ratio. Metabolites were annotated by
comparing observed m/z values with theoretical values in the Human
Metabolome Database v5.0 ([297]www.hmdb.ca), and m/z annotations were
assigned if errors fell within 10 ppm. To ensure the precision of the
annotations, metabolites underwent further identification employing
MS/MS. For metabolites involved in treating OS cells and worms
including hypotaurine, phosphocreatine, and O-phosphoethanolamine,
their confirmation relied on matching MS/MS spectra between cells and
standards (Supplementary Fig. [298]17). Other metabolites were
identified by comparing MS/MS spectra between bulk cellular samples and
the HMDB database. Batch effects of experiments were assessed through
PCA and HCA analysis in the R statistical environment.
Flow cytometry
The cells were seeded and cultured until they achieved a confluency of
70-80%. Following a single wash with 1 mL of phosphate-buffered saline
(PBS), the cells were dissociated using 0.25% trypsin solution
containing EDTA, after which the reaction was halted by supplementing
DMEM with FBS at a concentration of 10%. Subsequently, the cell
suspension was collected in tubes measuring approximately 1.5 mL
capacity and subjected to centrifugation at a speed of 600 × g for 5
min before being resuspended in PBS containing mClB at a final
concentration of 40 μM. The resuspended cells were then incubated at a
temperature of precisely maintained at or around 37 °C for 20 min.
Subsequently, the cells were washed and resuspended in PBS supplemented
with 1% FBS. After filtration through a 40 μm cell strainer and
transfer to clean tubes, all samples were vortexed for 5 s to ensure
complete dissociation into single-cell suspension prior to flow
cytometry analysis. Flow cytometric analysis was performed using a BD
FACSAria III instrument (BD Biosciences), with initial gating based on
FSC-A versus SSC-A parameters followed by measurement of whole-cell
fluorescence. Unstained cells were utilized as a blank control to
establish baseline correction. Finally, cells stained with mClB
exhibiting the top and bottom 5% fluorescent intensity were isolated
and collected independently for subsequent analysis. The FACS data was
then analyzed and visualized using the ‘fca_readfcs’ function and the
‘Flow cytometry GUI for MATLAB’ plugin in MATLAB (version 2022b)
([299]www.mathworks.com/matlabcentral/fileexchange/9608-fca_readfcs and
[300]www.mathworks.com/matlabcentral/fileexchange/38080-flow-cytometry-
gui-for-matlab).
Identification of marker metabolites
Wilcoxon rank-sum tests were used to compare metabolites in cells
belonging to one cluster with those of all other cells, based on the
results obtained from unsupervised clustering analysis. Ions exhibiting
P < 0.05 and fold change (FC) > 1.5 were identified as marker
metabolites for the specific cluster, which were subsequently z score
scaled and visualized using MATLAB’s ‘heatmap’ function. The z score
scales the data for each ion and is calculated as follows:
[MATH: z=x−X¯S :MATH]
1
where z represents z score, x represents a sample raw data,
[MATH: X¯
:MATH]
represents the population mean, and S represents the population
standard deviation.
Identification of metabolic subtypes
The metabolic data matrix was imported into the MATLAB workspace, where
it underwent z score scaling and unsupervised clustering using the
k-medoids algorithm. The resulting clusters were visualized with
Uniform Manifold Approximation and Projection (UMAP) via a MATLAB
plugin
([301]https://www.mathworks.com/matlabcentral/fileexchange/71902). The
data was dimensionally reduced to two dimensions and subsequently
visualized using the ‘gscatter’ function in MATLAB (version 2022b),
revealing distinct metabolic subtypes of cells within a bi-dimensional
space.
Pseudotime analysis and single-cell trajectory construction
The metabolite data matrix was initially filtered based on the
correlation with oxidative stress level (indicated by DCFDA intensity).
Metabolites exhibiting a correlation coefficient (r) greater than 0.2
or less than -0.2 and P < 0.05 in correlation analysis were selected
for pseudotime analysis. The refined data matrix was then imported into
Monocle (version 2.16.0) in R statistical environment, followed by
dimensional reduction using the ‘DDRTree’ algorithm according to the
documentation of Monocle
([302]https://cole-trapnell-lab.github.io/monocle-release/). Finally,
the ‘plot_cell_trajectory’ function was utilized to visualize the
trajectory.
Supervised machine learning and model evaluation
Supervised machine learning was conducted using MATLAB (version 2022b).
The data matrix was randomly divided into a training dataset and a
testing dataset at a ratio of 2:1. The testing dataset was exclusively
used for evaluating the trained model and not exposed to the training
phase. The training of both the classification and regression models
was conducted without any feature selection, and all m/z signals that
met our criteria (S/N > 3 and detected in greater than 20% single
cells) were included in both the training and testing datasets. To
train classification models, ensemble algorithm (function
‘fitcensemble’), discriminant analysis algorithm (function ‘fitcdiscr’)
and neural network algorithm (function ‘fitcnet’) were employed as
specified. The model underwent 5-fold cross-validation during training,
with Bayesian optimization employed to optimize hyperparameters and
minimize cross-validation loss (error). Parameters ‘Method’,
‘NumLearningCycles’, ‘MinLeafSize’, and ‘LearnRate’ were automatically
optimized in the ensemble algorithm, while parameters ‘Delta’ and
‘Gamma’ were automatically optimized in the discriminant analysis
algorithm. Parameters such as ‘Activations’, ‘Lambda’, ‘LayerSizes’,
and ‘Standardize’ were automatically optimized for neural network
algorithm. For the training of regression models, we utilized the
neural network algorithm (function ‘fitrnet’). The data underwent a
“log2” or “ln” transformation and was trained using 5-fold
cross-validation and Bayesian optimization. Parameters such as
‘Activations’, ‘Lambda’, ‘LayerSizes’, and ‘Standardize’ were
automatically optimized.
After training the model with the training dataset, the testing dataset
was utilized to evaluate the performance of the classification models
through receiver operating characteristic (ROC) curve and confusion
matrix analysis. For multiclass problem, ‘one versus rest’ strategy was
used which transformed the problem into a two-classification task. The
predicted vs real scatter plot and the Pearson’s r and P value were
used to evaluate the performance of the regression model.
Evaluation of metabolic similarity
The ‘pdist’ function in MATLAB (version 2022b) was utilized to compute
the statistical distance based on the cellular metabolome, where a
larger distance indicated a lower degree of similarity between two
cells. Similarity was calculated as the reciprocal of distance. The
heatmap was generated by plotting the reciprocal of the distance as a
measure of similarity, with higher values indicating greater likelihood
of shared metabolomic features between cells. We quantified the
pairwise distances between cells in the initial and OS groups based on
their metabolite abundance as variables. Spearman distance functions
were employed.
Spearman distance function:
[MATH:
ds,t=1−rs−r¯srt−r¯t′rs−r¯srs−r¯s′rt−r¯trt−r¯t′ :MATH]
2
where d[s,t] represents the distance between two cells (s and t); r[sj]
is the rank of x[sj] taken over x[1j], x[2j],…x[mj]; r[s] and r[t] are
the coordinate-wise rank vectors of x[s] and x[t], as an example,
r[s] = (r[s1], r[s2],… r[sn]);
[MATH: r¯s=1
n∑i−1n<
mi>rsi :MATH]
and
[MATH: r¯t=1
n∑i−1n<
mi>rti :MATH]
. n represents the number of variables.
Hamming distance was used in the comparison of metabolome of Hypt, PCr,
O-PE treated OS cells with Non-OS and vehicle treated OS cells.
Hamming distance function:
[MATH:
ds,t=#xsj≠
mo>xtjn
:MATH]
3
where d[s,t] represents the distance between two cells (s and t); x[sj]
represents the variable j of the first cell s, x[tj] represents the
variable j of the second cell t; n represents the number of variables.
The function calculates the fraction of different variables in all
variables (n) of the two cells.
Metabolite set enrichment analysis
Metabolite set enrichment analysis (MSEA) was performed using
MetaboAnalyst (v6.0), an online metabolomics analysis tool
([303]www.metaboanalyst.ca). The annotation of metabolites, including
metabolic markers in clusters and those correlated with the oxidative
stress level of single cells, was compiled into a list and uploaded to
the website. Only metabolites identified with MS/MS confirmation was
used in the MSEA. The algorithm processed the data and obtained
enriched pathways, while also calculating two important parameters:
Enrichment Ratio and P value. Enrichment Ratio indicates the degree of
enrichment, while P value represents its significance. Pathways with a
P value less than 0.05 were deemed significant. The results were
downloaded from the website and visualized using MATLAB’s ‘bubblechart’
function.
Construction of metabolic networks and rewiring analysis
The annotated metabolite data was analyzed using the ‘corr’ function in
MATLAB (version 2022b), resulting in a matrix of correlation
coefficients (r). This matrix was then reorganized into three columns:
metabolite-1, metabolite-2, and their respective correlation
coefficient values. The reorganized matrix was imported into Cytoscape
(version 3.10.1) to construct metabolic networks based on the
correlations between metabolites. In analysis of Cluster-I and
Cluster-II in initial cells, metabolite pairs with P > 0.05 or
|r | <0.8 were excluded. In the network, nodes represent metabolites,
and edges represent correlations. The networks of correlation were
visualized using Cytoscape. By utilizing the DyNet algorithm to compare
two different networks, changes in relationships with other metabolites
for each individual metabolite were analyzed. Metabolite rewiring
scores were calculated using the DyNet algorithm and then visualized
through stem plot with MATLAB’s ‘stem’ function.
Statistical analysis
The statistical analysis was conducted using Microsoft Excel
(Microsoft), R statistical environment, MATLAB 2022b (Mathworks), and
GraphPad Prism (version 8). The Wilcox rank sum test was employed to
determine the significance of two-grouped data, while one-way ANOVA or
two-way ANOVA were utilized for multi-group or multi-factor data,
respectively. Distribution analysis (using the “Rayleigh” probability
density function) and calculation of interquartile range (IQR) and
median absolute deviation (MAD) were performed using MATLAB R2022b
(MathWorks). Sample size was not predetermined by statistical methods
in this study, but rather based on previous experience. The number of
samples (n) is indicated in the figures or figure legends.
Reporting summary
Further information on research design is available in the [304]Nature
Portfolio Reporting Summary linked to this article.
Supplementary information
[305]Supplementary Information^ (31.7MB, pdf)
[306]Reporting Summary^ (450.3KB, pdf)
[307]Supplementary Data 1^ (13.1KB, xlsx)
[308]Supplementary Data 2^ (99.4KB, zip)
[309]Transparent Peer Review file^ (13.3MB, pdf)
Source data
[310]Source Data^ (4.9MB, zip)
Acknowledgements