Mol. Cells 2021; 44(3): 136-145
Published online March 29, 2021
https://doi.org/10.14348/molcells.2021.2239
© The Korean Society for Molecular and Cellular Biology
Correspondence to : kimchuna@kribb.re.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Senescent cells that gradually accumulate during aging are one of the leading causes of aging. While senolytics can improve aging in humans as well as mice by specifically eliminating senescent cells, the effect of the senolytics varies in different cell types, suggesting variations in senescence. Various factors can induce cellular senescence, and the rate of accumulation of senescent cells differ depending on the organ. In addition, since the heterogeneity is due to the spatiotemporal context of senescent cells, in vivo studies are needed to increase the understanding of senescent cells. Since current methods are often unable to distinguish senescent cells from other cells, efforts are being made to find markers commonly expressed in senescent cells using bulk RNA-sequencing. Moreover, single-cell RNA (scRNA) sequencing, which analyzes the transcripts of each cell, has been utilized to understand the in vivo characteristics of the rare senescent cells. Recently, transcriptomic cell atlases for each organ using this technology have been published in various species. Novel senescent cells that do not express previously established marker genes have been discovered in some organs. However, there is still insufficient information on senescent cells due to the limited throughput of the scRNA sequencing technology. Therefore, it is necessary to improve the throughput of the scRNA sequencing technology or develop a way to enrich the rare senescent cells. The in vivo senescent cell atlas that is established using rapidly developing single-cell technologies will contribute to the precise rejuvenation by specifically removing senescent cells in each tissue and individual.
Keywords aging, cellular senescence, heterogeneity, single-cell RNA sequencing, transcriptomics
Various types of damages, such as those in tissues, cells, and molecules, accumulate during aging and often lead to cellular senescence (Soares et al., 2014; Zhang et al., 2015). Senescent cells are secretory cells that are still metabolically active, although their cell cycle is stably stopped (Dörr et al., 2013). Senescent cells accumulate with age and have a significant impact on the health and longevity of an individual (Biran et al., 2017; Burd et al., 2013; Jeyapalan et al., 2007; Zhu et al., 2014). Cellular senescence is induced by various stresses. For example, telomere shortening occurs due to repetitive cell division, leading to a kind of senescence called replicative senescence, or Hayflick’s limit (Biran et al., 2017; Burd et al., 2013; Jeyapalan et al., 2007; Zhu et al., 2014). Other stresses, including the activation of oncogenes such as Ras, the inhibition of tumor suppressor genes such as p53, the accumulation of DNA damage, chromatin disruption, and reactive oxygen species, can trigger senescence.
However, cellular senescence can also occur in specific physiological conditions, such as development and wound healing (Demaria et al., 2014; Storer et al., 2013). Senescent cells affect the surrounding cells by secreting senescence-associated secretory phenotypes (SASPs), including cytokines, chemokines, growth factors, and matrix metalloproteases (Acosta et al., 2008; Coppé et al., 2008; Krtolica et al., 2001). Depending on the physiological context, SASPs can be involved in diverse mechanisms, such as immune cell recruitment, inflammation, and extracellular matrix (ECM) remodeling (Borodkina et al., 2018; Coppé et al., 2008; Xu et al., 2015). For example, SASPs can result in immune activation, growth arrest, and differentiation, processes that are essential for tissue restoration. SASPs can also result in cell growth, migration, and invasion, which have adverse effects on cancer. This context difference also exists in young and old tissues. In young tissues, the effects of SASPs are temporary because senescent cells can be eliminated by the immune cells recruited by pro-inflammatory SASPs (Demaria et al., 2014; Kang et al., 2011; Xue et al., 2007); thus, senescent cells facilitate wound healing, tumor suppression, and the restoration of tissue homeostasis (Campisi, 2001; Demaria et al., 2014; Storer et al., 2013). Conversely, as we get older, the removal of senescent cells becomes slower, resulting in the gradual accumulation of senescent cells. In other words, the effect of SASPs persists, likely resulting in chronic inflammation, aging-related deterioration or disease, and tumorigenesis (Chen et al., 2020; Coppé et al., 2008; Krizhanovsky et al., 2008; Nathan and Ding, 2010). It remains unclear whether the accumulation of senescent cells inhibits the immune surveillance system or if the aging of the immune surveillance system leads to the accumulation of senescent cells (Ray and Yung, 2018).
While senescence has a beneficial aspect in facilitating development and repairing damage, it has another aspect that induces organ dysfunction and aging. Therefore, there have been various attempts to reverse aging by regulating the accumulation of senescent cells and SASPs (Paez‐Ribes et al., 2019). Among these efforts, senolytics attempt to improve aging by selectively eliminating senescent cells. For example, when
However, this strategy does not work effectively against all types of senescent cells since cellular senescence is induced by various mechanisms to have different transcription signatures (Hernandez-Segura et al., 2017). In particular, p16, a marker for senescent cells, is not only universally expressed in various senescent cell types but is also expressed after the cell cycle is temporarily stopped (Marthandan et al., 2014). In addition, the representative senolytic drugs, such as dasatinib, quercetin, and navitoclax, had different effects depending on the cell type (Zhu et al., 2015; 2016). These data suggest that some degree of heterogeneity exists in senescent cells. Therefore, it would be essential to consider the heterogeneity of senescent cells to eliminate senescent cells more efficiently.
When irreparable DNA damage occurs in a cell, it dies through mechanisms such as apoptosis or necrosis or activates senescence (Childs et al., 2014). The cell cycle of senescent cells is arrested through the DNA-damage-response dependent p53/p21 pathway or the p16INK4a/RB pathway (Serrano et al., 1993). The factors determining the choice between apoptosis and senescence remain elusive. Still, given that the pro-apoptotic factors
Various methods have been developed to detect senescent cells based on their characteristics. The SA-β-gal staining assay and quantitative analysis of biomarkers, such as p16, p21, and p53, are commonly used (Dimri et al., 1995; Krishnamurthy et al., 2004; Ressler et al., 2006; Sharpless and Sherr, 2015). However, these methods also detect non-senescent cells. For example, both quiescent cells, which are induced by serum starvation or pH change in a culture environment, and confluent cells are also stained positive in the SA-β gal staining assay (Yang and Hu, 2005). In addition,
Several studies have been tried to identify common markers observed in all senescent cells through unbiased transcriptomic
In addition, in another independently performed transcriptomic analysis, universally expressed genes were found in various cell lines, such as lung fibroblast, umbilical vein endothelial, and alveolar endothelial cells, using various cellular senescence induction stimuli, such as replicative, oncogene-induced, ionizing radiation-induced, or doxorubicin-induced senescence. According to this analysis, the expression level of senescence markers, such as
Interestingly, bulk RNA expression was found to be highly correlated to the cell type more than the method of inducing senescence. In other words, regardless of the senescence inducer, such as telomere shortening, ionizing radiation, doxorubicin treatment, and the epxression of the HRASG12V oncogene, the endothelial cells (HUVECs, HAECs) and fibroblasts (WI-38, IMR-90) are grouped, respectively (Casella et al., 2019). This observation suggests that cellular senescence is likely cell-type-specific. Furthermore, even within the same cell type, a transcriptomic feature related to the origin of cell types is more prominent than the senescence-related transcriptomic changes. These results suggest that the
Two main factors likely lead to the
Second, senescent cells are not static; their transcriptomic and epigenomic states may change over time (Fig. 2). By tracking transcriptomic changes over time, the senescence induced by ionizing radiation was divided into three stages: early senescence, after 4 days of senescence induction; intermediate senescence, after 10 days of senescence induction; and late senescence, after 20 days of senescence induction. The early, intermediate, and late stages were enriched for transcript related to DNA damage responses, signaling mediated by p38-MAPK, and cell cycle arrest, respectively. In addition, the SASP-encoding genes also significantly changed over time (Hernandez-Segura et al., 2017). Therefore, studies that consider the effects of temporal and spatial context are required to understand
Since senescent cells rarely exist in organs, it has been challenging to track and analyze their transcription signatures
Studies based on bulk RNA-sequencing have been used to track the longitudinal changes in transcriptomes in various mice tissues and expand the understanding of
It is unclear whether the transcriptomic changes in each organ during aging are due to a cell-intrinsic change or a change in cellular composition. Indeed, tracking changes in the cell populations, such as the accumulation of senescent cells, the infiltration of immune cells, and the reduction of stem cells, which can significantly influence changes in aging transcriptome, are challenging in bulk RNA-sequencing. Single-cell RNA-sequencing (scRNA-seq) technology, which can identify intra-organ and inter-organ heterogeneity by tracking changes in the transcriptome of each individual cell in the aging process, could be applied to aging research (Fig. 3).
Unlike bulk RNA-seq, scRNA-seq needs to distinguish every single cell and molecule from a mixed sample. Most scRNA-seq methods enrich the mRNA using poly(T) oligonucleotides with barcodes that can tag distinct cells and molecules. Specifically, the cell barcode identifies each cell to facilitate multiplexing. The unique molecule identifier (UMI) is a barcode that can identify each molecule, thereby correcting amplification biases. Meanwhile, template switching (TS) is used in the reverse transcription step to obtain full-length transcripts. When a cDNA molecule reaches the 5′-end of an mRNA molecule, a few nucleotides are added to the 3′-end of the cDNA by a specific reverse transcriptase. Then, a unique TS oligo binds to this site and uses it as a template, so that cDNA synthesis continues to the 5′-end of TS oligo. Afterward, the cDNA molecule is amplified by polymerase chain reaction or
Various microfluidic- and plate-based methods have been rapidly developed based on this process. For example, plate-based methods, such as Smart-seq2, STRT-seq, and Quartz-seq, have the advantage of higher cell capture rate and are applied in full-length or 3′- or 5′-end transcript sequencing. On the other hand, InDrops and Drop-seq, which are microfluidic system-based on single-cell encapsulation in droplets, can easily scale up the number of cells, thus increasing the possibility of detecting rare cells (Lafzi et al., 2018). Since each method has strengths and weaknesses, the appropriate method should be selected while considering the cell type, costs, efficiency, and scalability.
Recently, the
However, scRNA-seq has difficulty detecting senescent cells, which account for only 2% of specific
Due to ethical issues, scRNA-seq analysis has been performed only in a limited number of human tissues (Table 4 and references therein). In human lung tissues obtained from donors and pulmonary fibrosis patients, rare cell populations, such as senescent cells, appeared when pulmonary fibrosis progressed with age (Reyfman et al., 2019). Interestingly, senescence markers such as
Senescent cells, which have heterogeneous transcriptomic signature, are accumulated with aging. Moreover, the senescent cells can affect the surrounding cells and the whole body by SASPs. We need to understand not only the features but also the ecosystem of the senescent cells. However, the throughput of scRNA-seq, which has only captured approximately 10,000 cells to date, needs to be improved to capture the rare population of senescent cells thoroughly (Fig. 3). Also, a technology that enriches senescent cells needs to be developed. In addition, it will be necessary to establish an organ-specific,
Furthermore, the degree of heterogeneity of senescent cells
A detailed understanding of senescent cells
This work was supported by the National Research Foundation of Korea (NRF-2020R1C1C101220611) and KRIBB Research Initiative Program.
S.K. and C.K. wrote the manuscript.
The authors have no potential conflicts of interest to disclose.
List of commonly changed genes in senescent cells
Ensembl ID | Gene symbol | Status | Description | GO: Cellular component | Cell type | No. of RNA-seq |
---|---|---|---|---|---|---|
ENSG00000033100 | CHPF2 | Up | Chondroitin polymerizing factor 2 | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000083444 | PLOD1 | Up | Procollagen-lysine,2-oxoglutarate 5-dioxygenase 1 | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000084444 | FAM234B | Up | Family with sequence similarity 234 member B | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000112697 | TMEM30A | Up | Transmembrane protein 30A | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000186866 | POFUT2 | Up | Protein O-fucosyltransferase 2 | Endoplasmic reticulum membrane | Fibroblast, endothelial cells, melanocytes, keratinocytes, astrocytes | 20 |
ENSG00000197077 | KIAA1671 | Up | Unknown protein coding | Unknown | Fibroblast, endothelial cells | 17 |
ENSG00000143815 | LBR | Down | Lamin B receptor | Membrane | Fibroblast, endothelial cells | 17 |
Fibroblast cells: WI-38, IMR90, HCA-2, BJ, HFF, MRC-5. Endothelial cells: HUVEC, HAEC.
Multiple cell types bulk RNA-seq studies (
Hernandez-Segura et al., (2017) | Casella et al., (2019) | |
---|---|---|
Cell lines | 6 different fibroblast strains (BJ, IMR90, HFF, MRC5, WI38, and HCA2), human neonatal foreskin epidermal melanocytes, keratinocytes, human fetal astrocytes | Human diploid fibroblast from fetal lung (WI-38, IMR-90), human aortic endothelial cells (HAECs), human umbilical vein endothelial cells (HUVECs) |
Induction stimuli | RS, OIS, IRIS, oxidative stress | RS, IRIS, OIS, doxorubicin |
Platform | Illumina Hiseq 2000 | Illumina Hiseq 4000 |
No. of common DEGs | 55 | 68 |
p16 in DEGs | No | No |
RS, replicative senescence; OIS, oncogene-induced senescence; IRIS, ionic radiation-induced senescence.
Multiple cell types scRNA-seq studies (
Kimmel et al., (2019) | Almanzar et al. (2020) | |
---|---|---|
Tissue | Kidney, lung, spleen | Bladder, bone marrow, brain (cerebellum, cortex, hippocampus and striatum), fat (brown, gonadal, mesenteric and subcutaneous), heart and aorta, kidney, large intestine, limb muscle and diaphragm, liver, lung, mammary gland, pancreas, skin, spleen, thymus, tongue and trachea |
Mice age (mo) | 7, 22-23 | 1, 3, 18, 21, 24, 30 |
Method | 10× Genomics | 10× Genomics, Smart-seq2 |
Capture format | Droplets | Droplets, plate |
Transcript coverage | 3’ end | 3’ end Full length |
No. of cells | 55,293 | 529,823 |
Single organ scRNA-seq studies (
Enge et al. (2017) | Hammond et al. (2019) | Yi et al. (2020) | Zheng et al. (2020) | De Micheli et al. (2020) | Zou et al. (2021) | |
---|---|---|---|---|---|---|
Species | Human | Mouse | Human, primate | Human | Human | Human |
Organ | Pancreas | Brain (microglia) | Retina | Peripheral blood mononuclear cells (PBMCs) | Muscle | Eyelid skin |
Age | Juvenile (1 mo, 5 y, 6 y), young adult (21 y, 22 y), adult/middle aged (38 y, 44 y, 54 y) | Embryonic day 14.5 (E14.5), early postnatal day 4/5 (P4/P5), late juvenile stage (P30), adulthood (P100), old age (P540), injury (P100) | Human: infant (8 days), adult (35-87 y) Macaque: juvenile (2 y), adults (4-23 y) | Cohort-1: young healthy adults (YA) (20-45 y), aged healthy adults (AA) (≥ 60 y) Cohort-2: comprising young adults (30-45 y) (YH), aged healthy adults (AH) (≥ 60 y), young COVID-19 onset patients (30-50 y), aged COVID-19 onset patients (≥ 70 y) Cohort-3: comprising YH AH, young recovered COVID-19 patients, aged recovered COVID-19 patients | Donors (range, 41-81 y) | Young, middle aged, old* |
Method | Smart-seq2 | 10× Genomics | 10× Genomics | 10× Genomics | 10× Genomics | 10× Genomics |
Capture format | Plate | Droplets | Droplets | Droplets | Droplets | Droplets |
Transcript coverage | Full length | 3’ end | 3’ end | 5’ end | 3’ end | 3’ end |
No. of cells | 2,544 | 76,149 | 119,520 | 166,609 | Over 22,000 | 35,678 |
Remark | An age-dependent mutational signature of endocrine cells is attributed to guanine oxidation selectively induced by reactive oxygen species. | Two microglia clusters are enriched in aging mice; one clustered has 2-4 times more microglia expressing inflammatory signals, such as Ccl4, Il1b, and Ccr5. | Human retinal aging occurs in the foveal region earlier. MYO9A− rods and the horizontal cell subtype, reduced in aging retina, are vulnerable to aging. | Age-induced immune cell polarization and expression of inflammation-related genes, such as FOS, DUSP1, IL1B, and cellular senescence-related genes, such as the CDKN family, are associated with vulnerability to COVID-19. | The muscle stem/progenitor cell (MuSC) population consists of MuSC1 and MuSC2 subpopulations. MuSC2 is enriched for inflammation markers, including CCL2, CXCL1, IL32, and TNFRSF12/FN14, that may constitute a marker set for MuSC variation in chronic muscle inflammation. | The cell-type specific downregulation of key TFs, such as KLF6 in keratinocytes and HES1 in dermal fibroblast, promote senescence phenotypes including increased SA-β-gal-positive cells and increased inflammation. |
*Specific ages are not defined.
Mol. Cells 2021; 44(3): 136-145
Published online March 31, 2021 https://doi.org/10.14348/molcells.2021.2239
Copyright © The Korean Society for Molecular and Cellular Biology.
Sohee Kim1,2 and Chuna Kim1, *
1Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea, 2Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
Correspondence to:kimchuna@kribb.re.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Senescent cells that gradually accumulate during aging are one of the leading causes of aging. While senolytics can improve aging in humans as well as mice by specifically eliminating senescent cells, the effect of the senolytics varies in different cell types, suggesting variations in senescence. Various factors can induce cellular senescence, and the rate of accumulation of senescent cells differ depending on the organ. In addition, since the heterogeneity is due to the spatiotemporal context of senescent cells, in vivo studies are needed to increase the understanding of senescent cells. Since current methods are often unable to distinguish senescent cells from other cells, efforts are being made to find markers commonly expressed in senescent cells using bulk RNA-sequencing. Moreover, single-cell RNA (scRNA) sequencing, which analyzes the transcripts of each cell, has been utilized to understand the in vivo characteristics of the rare senescent cells. Recently, transcriptomic cell atlases for each organ using this technology have been published in various species. Novel senescent cells that do not express previously established marker genes have been discovered in some organs. However, there is still insufficient information on senescent cells due to the limited throughput of the scRNA sequencing technology. Therefore, it is necessary to improve the throughput of the scRNA sequencing technology or develop a way to enrich the rare senescent cells. The in vivo senescent cell atlas that is established using rapidly developing single-cell technologies will contribute to the precise rejuvenation by specifically removing senescent cells in each tissue and individual.
Keywords: aging, cellular senescence, heterogeneity, single-cell RNA sequencing, transcriptomics
Various types of damages, such as those in tissues, cells, and molecules, accumulate during aging and often lead to cellular senescence (Soares et al., 2014; Zhang et al., 2015). Senescent cells are secretory cells that are still metabolically active, although their cell cycle is stably stopped (Dörr et al., 2013). Senescent cells accumulate with age and have a significant impact on the health and longevity of an individual (Biran et al., 2017; Burd et al., 2013; Jeyapalan et al., 2007; Zhu et al., 2014). Cellular senescence is induced by various stresses. For example, telomere shortening occurs due to repetitive cell division, leading to a kind of senescence called replicative senescence, or Hayflick’s limit (Biran et al., 2017; Burd et al., 2013; Jeyapalan et al., 2007; Zhu et al., 2014). Other stresses, including the activation of oncogenes such as Ras, the inhibition of tumor suppressor genes such as p53, the accumulation of DNA damage, chromatin disruption, and reactive oxygen species, can trigger senescence.
However, cellular senescence can also occur in specific physiological conditions, such as development and wound healing (Demaria et al., 2014; Storer et al., 2013). Senescent cells affect the surrounding cells by secreting senescence-associated secretory phenotypes (SASPs), including cytokines, chemokines, growth factors, and matrix metalloproteases (Acosta et al., 2008; Coppé et al., 2008; Krtolica et al., 2001). Depending on the physiological context, SASPs can be involved in diverse mechanisms, such as immune cell recruitment, inflammation, and extracellular matrix (ECM) remodeling (Borodkina et al., 2018; Coppé et al., 2008; Xu et al., 2015). For example, SASPs can result in immune activation, growth arrest, and differentiation, processes that are essential for tissue restoration. SASPs can also result in cell growth, migration, and invasion, which have adverse effects on cancer. This context difference also exists in young and old tissues. In young tissues, the effects of SASPs are temporary because senescent cells can be eliminated by the immune cells recruited by pro-inflammatory SASPs (Demaria et al., 2014; Kang et al., 2011; Xue et al., 2007); thus, senescent cells facilitate wound healing, tumor suppression, and the restoration of tissue homeostasis (Campisi, 2001; Demaria et al., 2014; Storer et al., 2013). Conversely, as we get older, the removal of senescent cells becomes slower, resulting in the gradual accumulation of senescent cells. In other words, the effect of SASPs persists, likely resulting in chronic inflammation, aging-related deterioration or disease, and tumorigenesis (Chen et al., 2020; Coppé et al., 2008; Krizhanovsky et al., 2008; Nathan and Ding, 2010). It remains unclear whether the accumulation of senescent cells inhibits the immune surveillance system or if the aging of the immune surveillance system leads to the accumulation of senescent cells (Ray and Yung, 2018).
While senescence has a beneficial aspect in facilitating development and repairing damage, it has another aspect that induces organ dysfunction and aging. Therefore, there have been various attempts to reverse aging by regulating the accumulation of senescent cells and SASPs (Paez‐Ribes et al., 2019). Among these efforts, senolytics attempt to improve aging by selectively eliminating senescent cells. For example, when
However, this strategy does not work effectively against all types of senescent cells since cellular senescence is induced by various mechanisms to have different transcription signatures (Hernandez-Segura et al., 2017). In particular, p16, a marker for senescent cells, is not only universally expressed in various senescent cell types but is also expressed after the cell cycle is temporarily stopped (Marthandan et al., 2014). In addition, the representative senolytic drugs, such as dasatinib, quercetin, and navitoclax, had different effects depending on the cell type (Zhu et al., 2015; 2016). These data suggest that some degree of heterogeneity exists in senescent cells. Therefore, it would be essential to consider the heterogeneity of senescent cells to eliminate senescent cells more efficiently.
When irreparable DNA damage occurs in a cell, it dies through mechanisms such as apoptosis or necrosis or activates senescence (Childs et al., 2014). The cell cycle of senescent cells is arrested through the DNA-damage-response dependent p53/p21 pathway or the p16INK4a/RB pathway (Serrano et al., 1993). The factors determining the choice between apoptosis and senescence remain elusive. Still, given that the pro-apoptotic factors
Various methods have been developed to detect senescent cells based on their characteristics. The SA-β-gal staining assay and quantitative analysis of biomarkers, such as p16, p21, and p53, are commonly used (Dimri et al., 1995; Krishnamurthy et al., 2004; Ressler et al., 2006; Sharpless and Sherr, 2015). However, these methods also detect non-senescent cells. For example, both quiescent cells, which are induced by serum starvation or pH change in a culture environment, and confluent cells are also stained positive in the SA-β gal staining assay (Yang and Hu, 2005). In addition,
Several studies have been tried to identify common markers observed in all senescent cells through unbiased transcriptomic
In addition, in another independently performed transcriptomic analysis, universally expressed genes were found in various cell lines, such as lung fibroblast, umbilical vein endothelial, and alveolar endothelial cells, using various cellular senescence induction stimuli, such as replicative, oncogene-induced, ionizing radiation-induced, or doxorubicin-induced senescence. According to this analysis, the expression level of senescence markers, such as
Interestingly, bulk RNA expression was found to be highly correlated to the cell type more than the method of inducing senescence. In other words, regardless of the senescence inducer, such as telomere shortening, ionizing radiation, doxorubicin treatment, and the epxression of the HRASG12V oncogene, the endothelial cells (HUVECs, HAECs) and fibroblasts (WI-38, IMR-90) are grouped, respectively (Casella et al., 2019). This observation suggests that cellular senescence is likely cell-type-specific. Furthermore, even within the same cell type, a transcriptomic feature related to the origin of cell types is more prominent than the senescence-related transcriptomic changes. These results suggest that the
Two main factors likely lead to the
Second, senescent cells are not static; their transcriptomic and epigenomic states may change over time (Fig. 2). By tracking transcriptomic changes over time, the senescence induced by ionizing radiation was divided into three stages: early senescence, after 4 days of senescence induction; intermediate senescence, after 10 days of senescence induction; and late senescence, after 20 days of senescence induction. The early, intermediate, and late stages were enriched for transcript related to DNA damage responses, signaling mediated by p38-MAPK, and cell cycle arrest, respectively. In addition, the SASP-encoding genes also significantly changed over time (Hernandez-Segura et al., 2017). Therefore, studies that consider the effects of temporal and spatial context are required to understand
Since senescent cells rarely exist in organs, it has been challenging to track and analyze their transcription signatures
Studies based on bulk RNA-sequencing have been used to track the longitudinal changes in transcriptomes in various mice tissues and expand the understanding of
It is unclear whether the transcriptomic changes in each organ during aging are due to a cell-intrinsic change or a change in cellular composition. Indeed, tracking changes in the cell populations, such as the accumulation of senescent cells, the infiltration of immune cells, and the reduction of stem cells, which can significantly influence changes in aging transcriptome, are challenging in bulk RNA-sequencing. Single-cell RNA-sequencing (scRNA-seq) technology, which can identify intra-organ and inter-organ heterogeneity by tracking changes in the transcriptome of each individual cell in the aging process, could be applied to aging research (Fig. 3).
Unlike bulk RNA-seq, scRNA-seq needs to distinguish every single cell and molecule from a mixed sample. Most scRNA-seq methods enrich the mRNA using poly(T) oligonucleotides with barcodes that can tag distinct cells and molecules. Specifically, the cell barcode identifies each cell to facilitate multiplexing. The unique molecule identifier (UMI) is a barcode that can identify each molecule, thereby correcting amplification biases. Meanwhile, template switching (TS) is used in the reverse transcription step to obtain full-length transcripts. When a cDNA molecule reaches the 5′-end of an mRNA molecule, a few nucleotides are added to the 3′-end of the cDNA by a specific reverse transcriptase. Then, a unique TS oligo binds to this site and uses it as a template, so that cDNA synthesis continues to the 5′-end of TS oligo. Afterward, the cDNA molecule is amplified by polymerase chain reaction or
Various microfluidic- and plate-based methods have been rapidly developed based on this process. For example, plate-based methods, such as Smart-seq2, STRT-seq, and Quartz-seq, have the advantage of higher cell capture rate and are applied in full-length or 3′- or 5′-end transcript sequencing. On the other hand, InDrops and Drop-seq, which are microfluidic system-based on single-cell encapsulation in droplets, can easily scale up the number of cells, thus increasing the possibility of detecting rare cells (Lafzi et al., 2018). Since each method has strengths and weaknesses, the appropriate method should be selected while considering the cell type, costs, efficiency, and scalability.
Recently, the
However, scRNA-seq has difficulty detecting senescent cells, which account for only 2% of specific
Due to ethical issues, scRNA-seq analysis has been performed only in a limited number of human tissues (Table 4 and references therein). In human lung tissues obtained from donors and pulmonary fibrosis patients, rare cell populations, such as senescent cells, appeared when pulmonary fibrosis progressed with age (Reyfman et al., 2019). Interestingly, senescence markers such as
Senescent cells, which have heterogeneous transcriptomic signature, are accumulated with aging. Moreover, the senescent cells can affect the surrounding cells and the whole body by SASPs. We need to understand not only the features but also the ecosystem of the senescent cells. However, the throughput of scRNA-seq, which has only captured approximately 10,000 cells to date, needs to be improved to capture the rare population of senescent cells thoroughly (Fig. 3). Also, a technology that enriches senescent cells needs to be developed. In addition, it will be necessary to establish an organ-specific,
Furthermore, the degree of heterogeneity of senescent cells
A detailed understanding of senescent cells
This work was supported by the National Research Foundation of Korea (NRF-2020R1C1C101220611) and KRIBB Research Initiative Program.
S.K. and C.K. wrote the manuscript.
The authors have no potential conflicts of interest to disclose.
. List of commonly changed genes in senescent cells.
Ensembl ID | Gene symbol | Status | Description | GO: Cellular component | Cell type | No. of RNA-seq |
---|---|---|---|---|---|---|
ENSG00000033100 | CHPF2 | Up | Chondroitin polymerizing factor 2 | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000083444 | PLOD1 | Up | Procollagen-lysine,2-oxoglutarate 5-dioxygenase 1 | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000084444 | FAM234B | Up | Family with sequence similarity 234 member B | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000112697 | TMEM30A | Up | Transmembrane protein 30A | Membrane | Fibroblast, endothelial cells | 17 |
ENSG00000186866 | POFUT2 | Up | Protein O-fucosyltransferase 2 | Endoplasmic reticulum membrane | Fibroblast, endothelial cells, melanocytes, keratinocytes, astrocytes | 20 |
ENSG00000197077 | KIAA1671 | Up | Unknown protein coding | Unknown | Fibroblast, endothelial cells | 17 |
ENSG00000143815 | LBR | Down | Lamin B receptor | Membrane | Fibroblast, endothelial cells | 17 |
Fibroblast cells: WI-38, IMR90, HCA-2, BJ, HFF, MRC-5. Endothelial cells: HUVEC, HAEC..
. Multiple cell types bulk RNA-seq studies (
Hernandez-Segura et al., (2017) | Casella et al., (2019) | |
---|---|---|
Cell lines | 6 different fibroblast strains (BJ, IMR90, HFF, MRC5, WI38, and HCA2), human neonatal foreskin epidermal melanocytes, keratinocytes, human fetal astrocytes | Human diploid fibroblast from fetal lung (WI-38, IMR-90), human aortic endothelial cells (HAECs), human umbilical vein endothelial cells (HUVECs) |
Induction stimuli | RS, OIS, IRIS, oxidative stress | RS, IRIS, OIS, doxorubicin |
Platform | Illumina Hiseq 2000 | Illumina Hiseq 4000 |
No. of common DEGs | 55 | 68 |
p16 in DEGs | No | No |
RS, replicative senescence; OIS, oncogene-induced senescence; IRIS, ionic radiation-induced senescence..
. Multiple cell types scRNA-seq studies (
Kimmel et al., (2019) | Almanzar et al. (2020) | |
---|---|---|
Tissue | Kidney, lung, spleen | Bladder, bone marrow, brain (cerebellum, cortex, hippocampus and striatum), fat (brown, gonadal, mesenteric and subcutaneous), heart and aorta, kidney, large intestine, limb muscle and diaphragm, liver, lung, mammary gland, pancreas, skin, spleen, thymus, tongue and trachea |
Mice age (mo) | 7, 22-23 | 1, 3, 18, 21, 24, 30 |
Method | 10× Genomics | 10× Genomics, Smart-seq2 |
Capture format | Droplets | Droplets, plate |
Transcript coverage | 3’ end | 3’ end Full length |
No. of cells | 55,293 | 529,823 |
. Single organ scRNA-seq studies (
Enge et al. (2017) | Hammond et al. (2019) | Yi et al. (2020) | Zheng et al. (2020) | De Micheli et al. (2020) | Zou et al. (2021) | |
---|---|---|---|---|---|---|
Species | Human | Mouse | Human, primate | Human | Human | Human |
Organ | Pancreas | Brain (microglia) | Retina | Peripheral blood mononuclear cells (PBMCs) | Muscle | Eyelid skin |
Age | Juvenile (1 mo, 5 y, 6 y), young adult (21 y, 22 y), adult/middle aged (38 y, 44 y, 54 y) | Embryonic day 14.5 (E14.5), early postnatal day 4/5 (P4/P5), late juvenile stage (P30), adulthood (P100), old age (P540), injury (P100) | Human: infant (8 days), adult (35-87 y) Macaque: juvenile (2 y), adults (4-23 y) | Cohort-1: young healthy adults (YA) (20-45 y), aged healthy adults (AA) (≥ 60 y) Cohort-2: comprising young adults (30-45 y) (YH), aged healthy adults (AH) (≥ 60 y), young COVID-19 onset patients (30-50 y), aged COVID-19 onset patients (≥ 70 y) Cohort-3: comprising YH AH, young recovered COVID-19 patients, aged recovered COVID-19 patients | Donors (range, 41-81 y) | Young, middle aged, old* |
Method | Smart-seq2 | 10× Genomics | 10× Genomics | 10× Genomics | 10× Genomics | 10× Genomics |
Capture format | Plate | Droplets | Droplets | Droplets | Droplets | Droplets |
Transcript coverage | Full length | 3’ end | 3’ end | 5’ end | 3’ end | 3’ end |
No. of cells | 2,544 | 76,149 | 119,520 | 166,609 | Over 22,000 | 35,678 |
Remark | An age-dependent mutational signature of endocrine cells is attributed to guanine oxidation selectively induced by reactive oxygen species. | Two microglia clusters are enriched in aging mice; one clustered has 2-4 times more microglia expressing inflammatory signals, such as Ccl4, Il1b, and Ccr5. | Human retinal aging occurs in the foveal region earlier. MYO9A− rods and the horizontal cell subtype, reduced in aging retina, are vulnerable to aging. | Age-induced immune cell polarization and expression of inflammation-related genes, such as FOS, DUSP1, IL1B, and cellular senescence-related genes, such as the CDKN family, are associated with vulnerability to COVID-19. | The muscle stem/progenitor cell (MuSC) population consists of MuSC1 and MuSC2 subpopulations. MuSC2 is enriched for inflammation markers, including CCL2, CXCL1, IL32, and TNFRSF12/FN14, that may constitute a marker set for MuSC variation in chronic muscle inflammation. | The cell-type specific downregulation of key TFs, such as KLF6 in keratinocytes and HES1 in dermal fibroblast, promote senescence phenotypes including increased SA-β-gal-positive cells and increased inflammation. |
*Specific ages are not defined..
Chanhee Kang
Mol. Cells 2019; 42(12): 821-827 https://doi.org/10.14348/molcells.2019.0298Yoojin Kwon, Ji Wook Kim, Jo Ae Jeoung, Mi-Sung Kim, and Chanhee Kang
Mol. Cells 2017; 40(9): 607-612 https://doi.org/10.14348/molcells.2017.0151Seon Beom Song, Woosung Shim, and Eun Seong Hwang*
Mol. Cells 2023; 46(8): 486-495 https://doi.org/10.14348/molcells.2023.0019