Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
Review Article50th Anniversary Celebration Collection Special Section on Drug Metabolism and Precision Medicine—Minireview
Open Access

Achieving a Deeper Understanding of Drug Metabolism and Responses Using Single-Cell Technologies

Abigail M. Wheeler, Colten D. Eberhard, Eric P. Mosher, Yuting Yuan, Hannah N. Wilkins, Herana Kamal Seneviratne, Benjamin C. Orsburn and Namandjé N. Bumpus
Drug Metabolism and Disposition March 2023, 51 (3) 350-359; DOI: https://doi.org/10.1124/dmd.122.001043
Abigail M. Wheeler
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Colten D. Eberhard
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eric P. Mosher
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Eric P. Mosher
Yuting Yuan
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hannah N. Wilkins
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Herana Kamal Seneviratne
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Herana Kamal Seneviratne
Benjamin C. Orsburn
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Namandjé N. Bumpus
Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland (A.M.W., C.D.E., E.P.M., Y.Y., H.N.W., H.K.S., B.C.O., N.N.B.) and Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland (H.K.S.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Recent advancements in single-cell technologies have enabled detection of RNA, proteins, metabolites, and xenobiotics in individual cells, and the application of these technologies has the potential to transform pharmacological research. Single-cell data has already resulted in the development of human and model species cell atlases, identifying different cell types within a tissue, further facilitating the characterization of tumor heterogeneity, and providing insight into treatment resistance. Research discussed in this review demonstrates that distinct cell populations express drug metabolizing enzymes to different extents, indicating there may be variability in drug metabolism not only between organs, but within tissue types. Additionally, we put forth the concept that single-cell analyses can be used to expose underlying variability in cellular response to drugs, providing a unique examination of drug efficacy, toxicity, and metabolism. We will outline several of these techniques: single-cell RNA-sequencing and mass cytometry to characterize and distinguish different cell types, single-cell proteomics to quantify drug metabolizing enzymes and characterize cellular responses to drug, capillary electrophoresis-ultrasensitive laser-induced fluorescence detection and single-probe single-cell mass spectrometry for detection of drugs, and others. Emerging single-cell technologies such as these can comprehensively characterize heterogeneity in both cell-type-specific drug metabolism and response to treatment, enhancing progress toward personalized and precision medicine.

SIGNIFICANCE STATEMENT Recent technological advances have enabled the analysis of gene expression and protein levels in single cells. These types of analyses are important to investigating mechanisms that cannot be elucidated on a bulk level, primarily due to the variability of cell populations within biological systems. Here, we summarize cell-type-specific drug metabolism and how pharmacologists can utilize single-cell approaches to obtain a comprehensive understanding of drug metabolism and cellular heterogeneity in response to drugs.

Introduction

In terms of morphology, function, and gene expression profiles, cell-to-cell differences are evident between various tissues, and have also been observed among the same cell types (Altschuler and Wu, 2010; Paszek et al., 2010). Paszek et al. proposed that cellular heterogeneity and coordination of signaling and regulatory systems, such as the cell cycle and apoptosis, allow a tissue as a whole to maintain biologic stability (Paszek et al., 2010). In recent years, a variety of comprehensive single-cell transcriptomic and genomic studies have demonstrated that heterogeneity is intrinsic to the nature of individual cells and arises from stochastic influences, such as the microenvironment (Peng et al., 2019; Izar et al., 2020). It is no longer a question of whether this heterogeneity exists, but rather how it affects our understanding of cellular function, metabolism, and even response to therapeutics.

Longstanding techniques involving the use of bulk cells or homogenized tissue only allow researchers to measure an average response of all the components of their sample. This type of measurement may not fully reflect physiologic differences between subpopulations or individual cells. As a means to address this, recent advances in single-cell technologies have enabled the use of multi-omics approaches to obtain a more comprehensive understanding of disease biology on a single-cell level. For example, analyzing a biopsy lysate prohibits the characterization of the tumor microenvironment at a cellular level; however, single-cell proteomics, epigenetic analyses, and a number of sequencing techniques – whole genome, whole exome, and single-cell RNA – have demonstrated the complexity of the tumor microenvironment (Wilting and Dannenberg, 2012; Gay et al., 2016; Slavov, 2022). This may contribute significantly to the development of resistance to anti-tumor treatment (Wilting and Dannenberg, 2012; Gay et al., 2016; Slavov, 2022). Thus, analyses of tumor biopsies fail to capture the regional heterogeneity of the tumor itself, potentially missing lesions essential for an accurate prognosis (Gay et al., 2016). It can be envisioned that single-cell technologies could be used to advance the understanding of intracellular pharmacokinetics and inter-cellular variability in drug response.

Here, we look forward and describe how techniques, such as single-cell RNA sequencing (scRNA-seq), a technology that can be enhanced by Drop-seq and mass cytometry, can be used to characterize different cell populations and investigate the expression of drug metabolizing enzymes in individual cells of specific cell populations. Methods that allow the visualization and analysis of cell-to-cell heterogeneity include co-detection by indexing, mass spectrometry-based proteomics and imaging, and whole-cell patch clamp electrophysiology paired with high-resolution mass spectrometry. Additionally, we will discuss opportunities for applying technologies, such as secondary ion mass spectrometry, single-probe single-cell mass spectrometry, and capillary electrophoresis-ultrasensitive laser-induced fluorescence detection to quantify and locate drugs in individual cells. In this review, we will cover the current knowledge pertaining to organ and cell-type-specific drug metabolism and propose how emerging single-cell technologies can be employed to expand the scope of the field of pharmacology and drug metabolism.

Cell-Type-Specific Drug Metabolism

Liver

Primary human hepatocytes are a commonly used model for investigating drug metabolism, as this system is often reflective of in vivo hepatic metabolism (Ponsoda et al., 2001; McGinnity et al., 2004). However, hepatocytes only make up approximately 80% of cells in the liver. Other cell types, including hepatic stellate cells, endothelial cells, cholangiocytes, and Kupffer cells, may also influence drug metabolism (MacParland et al., 2018; Aizarani et al., 2019). Importantly, these cells make up the complex framework of the liver and express a degree of intrapopulation heterogeneity themselves, which enables the multifunctionality of the organ (MacParland et al., 2018; Marcu et al., 2018; Payen et al., 2021).

Although a significant proportion of the body’s drug metabolism can be attributed to the liver, several other organs, including the kidneys, intestines, lungs, and brain, express metabolizing enzymes, such as the cytochromes P450 (P450s), UDP-glucuronosyltransferases (UGTs), sulfotransferases (SULTs), N-acetyltransferases (NATs), and others (Table 1). Notably, extrahepatic drug metabolizing enzymes can impact drug bioavailability and pharmacokinetics in both an organ and cell-type-specific manner (Paine et al., 1996; Zhang et al., 2007; Fonsi, 2014; Shibata and Chiba, 2015). For example, Shibata and Chiba observed significantly higher predicted bioavailability based on hepatocyte metabolism than calculated bioavailability in the plasma of rats, dogs, monkeys, and humans (Shibata and Chiba, 2015).

View this table:
  • View inline
  • View popup
TABLE 1

Summary of extrahepatic drug metabolizing enzymes

Kidney

Drug metabolism in the kidneys has been extensively studied and has been shown to contribute considerably to the biotransformation of a variety of different drug classes, including immunosuppressants, such as cyclosporin A, sedatives, such as propofol, and antiretrovirals, such as zidovudine (antiretroviral) (Dai et al., 2004; Hiraoka et al., 2005; Knights et al., 2016). Using scRNA-seq, ten distinct cell types were classified in adult human kidneys. Of these, glomerular parietal epithelial cells and proximal tubule cells accounted for 80% of the cells present (Liao et al., 2020). Primary human proximal tubule cells have been noted to express UGT1A1, UGT1A6, and UGT2B7, as well as CYP1B1, CYP3A4/5, and CYP4A11; however, the only enzymatic activity that was directly observed was that of CYP3A4/5 (Lash et al., 2008). UGTs 1A8/9 and 2B7 have been observed in primary renal epithelial and cortical renal epithelial cells, in addition to proximal tubule cells, but at significantly lower levels than in renal tissue as a whole (Van der Hauwaert et al., 2014). In immortalized cell lines, such as HK2 (proximal tubular cell line) and human embryonic kidney 293 cells, UGT transcript expression has not been detected (Van der Hauwaert et al., 2014). Differences between cellular and microsomal/tissue expression data suggest that there could be a range of cell types contributing to renal metabolism. These cells may be overlooked or underappreciated in bulk analyses; however, single-cell technologies have the potential to fill this gap when studying renal metabolism, as well as drug metabolism in other regions of the body.

Intestine

Drug-metabolizing enzymes are also expressed throughout the intestines, and have been found to contribute to the overall clearance of drugs, such as cyclosporin, midazolam, and docetaxel (Kolars et al., 1991; Thummel et al., 1996; van Herwaarden et al., 2007). RNA sequencing of human intestinal epithelial cells revealed the presence of several phase 1 and phase 2 drug metabolizing enzymes, with notable differences in transcript expression between the duodenum, ileum, colon, and rectum. In the ileum, the most abundant P450 transcripts observed were CYP3A4, CYP2C18, CYP3A5, and CYP2J2 (Takayama et al., 2021).

Enterocytes, the major subtype of intestinal epithelial cells, from the duodenum exhibit nonspecific UGT and SULT activity as well as CYP3A4, CYP2C9, CYP2C19, and CYP2J2 activity (Ho et al., 2017). Of the four intestinal regions examined, these enzymes were more highly expressed in the ileum and duodenum than in the colon or rectum. This region-specific expression also included UGTs and SULTs, where UGT8, UGT1A1, UGT2B7, UGT2A3, and SULT2A1, SULT1B1, SULT1A2, and SULT1A1 were the top four enzymes of each class identified in the ileum (Takayama et al., 2021). An orthogonal study employing immunoquantification in tissue from the ileum and duodenum identified many of the same SULTs, but at slightly different proportions, confirming that whole tissue analyses do not necessarily reflect the protein abundance in specific cell populations (Riches et al., 2009).

Intestinal cellular models, such as Caco-2 cells, have shown low transcript expression of CYP3A4, which can be artificially induced, but limited endogenous expression of drug metabolizing enzymes make it a poor model for exploring intestinal drug metabolism (Sergent et al., 2009; Li, 2020). Intestinal metabolism has also been explored using precision-cut intestinal slices as a more comprehensive approach than classic in vitro models, such as cultured cells or microsomes (de Graaf et al., 2010). In precision-cut intestinal slices taken from both the ileum and the colon, CYP3A and SULT1 metabolite formation was higher in the ileum, while CYP2A and UGT1A metabolite formation was higher in the colon, indicating regional variability (Iswandana et al., 2018). However, this method does not allow for the discernment between different cell types. Understanding cell-type-specific metabolism within different intestinal regions is especially relevant for regimens that are delivered rectally or drugs that specifically target the intestines.

Lung

Regional differences in metabolizing enzymes have also been noted in the lung, another extrahepatic organ in which local metabolism may have significant effects on overall drug biotransformation, particularly for medications administered via inhalation. scRNA-seq has characterized 58 different cell types in the human lung, encompassing epithelial, endothelial, stromal, and immune cell populations (Travaglini et al., 2020).

Parenchymal cells, part of the alveolar epithelial population, have been found to express the following P450 enzymes: CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4 and CYP3A5, all at a fraction of the expression levels found in the hepatocytes. However, phase 2 metabolizing enzymes SULT1A1, SULT1A3/4, SULT1C2, SULT2B1/2, and UGT2A1 were expressed more highly in the parenchymal cells. Several other UGTs and SULT1A2 were also observed, but with lower relative expression to hepatocytes (Somers et al., 2007).

Further, primary human bronchial epithelial cells have been noted to express up to 27 different P450 isoforms, including CYP1A1, CYP2C9, CYP2J2, and CYP3A5, which were also observed in alveolar epithelial cells (Courcot et al., 2012). Courcot et al. also presented phase 2 metabolizing enzyme mRNA expression of UGTs 1A1 and 1A6, NAT5, and SULTs 1A3/4, 1B1, 1E1, 2B1, and 4A1 (Courcot et al., 2012). Of particular interest, transcripts of P450s 1A1 and 1B1 have been observed at higher concentrations in the lungs of smokers and ex-smokers as opposed to non-smokers, indicating that environmental and/or health-related events could impact drug metabolism in the lung (Kim et al., 2004).

Brain

The brain is perhaps the most pharmacologically elusive and heterogenous of the organs discussed here. When considering the presence of drug metabolizing enzymes, there are not only several brain regions, but a multitude of cell types that must be taken into account. In the cortex alone, there have been 180 perceptible areas (Glasser et al., 2016) and 75 cell types identified (Hodge et al., 2019). Between the frontal cortex, striatum, hippocampus, thalamus/hypothalamus, basal ganglia, substantia nigra, amygdala, pons, and cerebellum, the frontal cortex and cerebellum are the most expressive of P450 enzymes, with 11 and 10 P450s identified, respectively, while the thalamus and hypothalamus have only two P450 enzymes expressed: CYP2D6 and CYP3A4 (McFadyen et al., 1998; Toselli et al., 2015; Kuban and Daniel, 2021). Although CYP2B6 mRNA has been observed in five of the different brain regions explored (frontal cortex, striatum, basal ganglia, pons, and cerebellum), a study examining the microsomal metabolism of various CYP2B6 substrates — nicotine, ketamine, and methadone — found no evidence of enzyme activity (Bloom et al., 2019).

Importantly, CYP2E1 plays a role in the metabolism of ethanol in the brain (Carpenter et al., 1996; Upadhya et al., 2000; Zimatkin et al., 2006; Zhong et al., 2012; Toselli et al., 2015) and has also been linked to Parkinson’s disease (Shahabi et al., 2008; Kaut et al., 2012). CYP2E1 mRNA is found primarily in neurons, but has also been observed in Purkinje cells of the cerebellum (Upadhya et al., 2000). In a study of the blood-brain barrier (BBB), CYP1A1, CYP1B1, CYP2A6, CYP2B6, CYP2C9, CYP2E1, CYP2J2, CYP3A4, and CYP4A11 mRNA were observed in primary human brain endothelial cells, each at a significantly higher level than commercially available immortalized human brain microvascular cerebral endothelial cells and human-derived umbilical vein endothelial cells (Ghosh et al., 2010). Further study of human brain microvessels, however, revealed that CYP1B1 and CYP2U1 were the only two P450s able to be quantified at the protein level (Shawahna et al., 2011).

Regarding phase 2 metabolizing enzymes, UGTs and NATs were not observed in the microvasculature of the BBB, but SULT1A1 was expressed (Shawahna et al., 2011). UGT mRNA expressed in brain tissues includes UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2A3, UGT2B4, UGT2B7, UGT2B10, UGT2B11, UGT2B15, UGT2B17, and UGT2B28 (Court et al., 2012; Jones and Lazarus, 2014). Both UGT1A6 and UGT2B7 mRNA have been observed specifically in the cerebellum (King et al., 1999). UGT2B7, the enzyme responsible for the biotransformation of morphine (Coffman et al., 1997), showed activity in some, but not all, human brain microsomes tested in a study by Wahlstrom et al. (Wahlstrom et al., 1988). Lastly, the protein of SULTs 1A1 and 1A3 have been identified in neurons and glial cells from the hippocampus, cerebellum, and frontal, occipital, and temporal lobes (Salman et al., 2009). Overall, these data indicate the presence of drug metabolizing enzymes in specific regions of the brain, which is of particular relevance to local metabolism of drugs that cross the blood-brain barrier.

Moving Forward

While the liver is the primary site of metabolism of most drugs, extrahepatic organs, such as the kidneys and the intestines, have been shown to contribute to the overall clearance of certain drugs. Additionally, extrahepatic metabolism has the potential to impact pharmacology when target tissues exhibit local metabolism. Further, with the complexity of cellular heterogeneity in pathophysiological states, such as tumorigenesis and the sensitivity of cell state to the microenvironment, investigation of cell-type-specific drug metabolism could reveal unique insight into cellular phenotype in these contexts (Wilting and Dannenberg, 2012; Kirschner et al., 2020; Guan et al., 2021; Xu et al., 2022). We envision the utilization of single-cell technologies in drug metabolism research will aid in more accurate targeting and dosing for a wide variety of treatments.

Current literature primarily uses scRNA-seq to address differences in the expression of drug metabolizing enzyme transcripts between different cell types, but this strategy is most powerful when employed in conjunction with other single-cell technologies. Few studies have sought to uncover intercellular heterogeneity of drug metabolizing enzymes in extrahepatic metabolizing organs, and the recent development of a variety of single-cell analysis tools will be indispensable in remedying this gap in knowledge.

Current and Emerging Single-Cell Methods

While some progress has been made using scRNA-seq to study drug-metabolizing enzyme transcript expression, there is still little known about drug metabolism and action in a single cell. The following methods can be employed to ascertain cell-type heterogeneity in metabolic activity and cellular response to aid in pharmacological research (Table 2).

View this table:
  • View inline
  • View popup
TABLE 2

Summary of single-cell methods

RNA Transcripts

Transcriptomics at the single-cell level has been used as a means to determine therapeutic targets and to identify genes related to drug tolerance. Drop-seq is a technique used to analyze mRNA of individual cells and has been applied to the investigation of genes associated with drug-tolerance (Macosko et al., 2015; Aissa et al., 2021). Using this method, Aissa et al. employed PC9 cells to define subpopulations of drug-tolerant cells after treatment with erlotinib, a tyrosine kinase inhibitor, and characterized previously unidentified drug-tolerant markers. In determining drug-tolerant markers in subpopulations, Aissa et al. identified drugs that could downregulate such tolerance markers, sensitizing drug-tolerant cells to erlotinib treatment, a feat not possible using classic bulk cell RNA sequencing technologies. Further, their data determined that higher transcript expression of tolerance markers correlated with worse patient outcomes (Aissa et al., 2021). Interestingly, drug tolerance was shown to be continual such that cells can transition between tolerance states, which is an important finding relevant to improving therapeutics. Overall, transcriptomics can be used to characterize subpopulation tolerances that can better inform personalized treatments, as well as identify new targets for drug discovery.

In recent years, bioinformaticians have generated various scRNA-seq atlases and databases, as the curation of datasets facilitates the continual analysis of published data. Several scRNA-seq databases have been established, such as the EMBL-EBI Single Cell Expression Atlas, which as of March 2022 includes 304 studies, encompassing 20 species and over 8.5 million cells (Moreno et al., 2022). Other initiatives, such as Gene Expression Omnibus (Barrett et al., 2009) and CellDepot (Lin et al., 2022), provide raw sequencing and visually processed data from single-cell sequencing data, respectively. In utilizing these databases, pharmacologists can uncover the transcript expression of drug-metabolizing enzymes in specific cell populations and identify potential therapeutic targets.

Proteins

While measurements of transcript abundance are valuable and have the potential to be integrated with other -omics data sets, single-cell protein abundance and localization data can further improve our understanding of complex biologic systems and lend insight into drug disposition. Of note, flow cytometry can be used to characterize heterogeneity in cell surface markers and intracellular proteins in many different biologic systems. However, this technique is limited in multiplexing capabilities and does not allow for characterization of protein localization within cells and intact tissues (Tracey et al., 2021).

Mass cytometry (also referred to as CyTOF) is a complementary method to flow cytometry that can produce comprehensive data sets. Like flow cytometry, CyTOF uses antibodies to cell surface markers and intracellular proteins to enable detection. However, while flow cytometry multiplexing is limited by spectral overlap of fluorophores, CyTOF uses antibodies conjugated to heavy metals and inductively coupled mass spectrometry to measure the abundance of these metals in each cell. This technique allows for routine 40-plex experiments and theoretically enables 100-plex studies.

Similar to scRNA-seq and flow cytometry, CyTOF has been used to study the immune tumor microenvironment (Norton et al., 2019; Zhang et al., 2019; Zhu et al., 2021). Zhu et al. were able to use such data to link AKT activity to chemotherapy resistance in breast cancer cells and showed that AKT inhibition increased cell death (Zhu et al., 2021). CyTOF provides a unique profile of a tumor and has been used to develop a computational framework to identify the optimal combination therapy for a patient (Anchang et al., 2018). Additionally, Zhang and colleagues incorporated CyTOF data into a multi-omics analysis including RNA-seq and metabolomics data to study the heterogeneity of hepatocellular carcinoma (Zhang et al., 2019).

The same antibodies that enable high multiplexing in CyTOF can also be used to build upon classic immunohistochemistry in imaging mass cytometry and multiplexed ion beam imaging (MIBI) experiments. Both methods involve labeling a tissue section with metal-conjugated antibodies and detecting these metals by mass spectrometry. To transfer the metals from the tissue to the mass spectrometer, imaging mass cytometry and MIBI use laser ablation and a beam of primary ions, respectively. Both techniques can achieve spatial resolution at or below 1 µm, rendering them capable of imaging at the single-cell level. Employing MIBI technology, Keren et al. characterized breast cancer tumor immune microenvironments using 36 markers simultaneously at 260 nm spatial resolution (Keren et al., 2019).

An emerging multiplexed protein imaging technique is co-detection by indexing (CODEX). CODEX utilizes oligonucleotide-conjugated antibodies and involves iterative in situ polymerization to selectively incorporate dNTP-tethered fluorophores on specific target barcodes. Following imaging, fluorophores are removed and additional rounds of in situ polymerization are completed, theoretically allowing for limitless protein detection. Goltsev and colleagues used CODEX to characterize the architecture of normal and diseased mouse spleens using 28 antibodies on the same tissue section (Goltsev et al., 2018).

Typically, these single-cell methods are used to compare the presence of surface markers and occasionally intracellular proteins. However, these methods can be used to inform knowledge of drug metabolism and pharmacokinetics by applying them to the study of drug transporters and intracellular drug metabolizing enzymes.

MS-based proteomics removes the need for cell surface markers to characterize proteins of a single cell. Combined with other biochemical methods, mass spectrometry can deliver data on regulatory correlation, signaling pathways, metabolic processes, and posttranslational modifications, all of which are factors that can drive cellular pathobiology and drug response (Olsen and Mann, 2013; Carvalho and Matthiesen, 2016; Marx, 2019; Ctortecka and Mechtler, 2021; Meissner et al., 2022).

The standard bottom-up proteomics workflow includes cell lysis, protein extraction and solubilization, reduction of disulfide bonds and alkylation of cysteines, digestion of proteins to peptides, sample desalting, clean-up, chromatographic separation, MS acquisition, and analysis (Aebersold and Mann, 2003; Bantscheff et al., 2007; Gundry et al., 2009). Currently, single cells are isolated by fluorescence-activated cell sorting or microscopy-based sorting techniques into multi-well plates, where lysis, digestion, and multiplex labeling, if employed, are performed. To date, multiple methods for single-cell proteomics processing have been described, including minimal proteomic sample preparation (Budnik et al., 2018), nanodroplet processing in one pot for trace samples (nanoPOTS) (Zhu et al., 2018), and Single Cell ProtEomics by Mass Spectrometry using the cellenONE automated single cell seeding platform (Vallone et al Rinke et al., 2014; Hartlmayr et al., 2021).

The nanoPOTS platform (Zhu et al., 2018) consists of microfabricated nanowell array chips and a nanoliter robotic pipetting system (Fig. 1A). The nanoPOTS platform demonstrated enhanced efficiency and sample recovery by downscaling processing volumes to < 200 µl to minimize surface losses (Zhu et al., 2018). Recent trials of the nanoPOTS platform have identified and quantified over 1,000 proteins from single mammalian cells (Kelly et al., 2019).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Three sample preparation strategies for single-cell proteomic analysis. (A) The nanoPOTS platform uses a nanoliter robotic pipetting to dispense cells and reagents onto microfabricated nanoarray chips (Zhu et al., 2018). (B) The ScoPE2 method uses TMT isobaric tags to label peptides of single cells and a 200-cell channel to boost protein sequence coverage (Petelski et al., 2021). (C) Whole-cell patch-clamp electrophysiology involves sample injection into a patch pipette, bottom-up proteomics on collected protein, and identification and quantification using a capillary electrophoresis electrospray ionization platform followed by high-resolution mass spectrometry (Choi et al., 2021).

Modern proteomics often relies on the use of chemical “barcodes” to facilitate sample multiplexing, allowing up to an 18-fold increase in sample throughput. Specifically, isobaric tandem mass tags (TMTs) have an identical total mass while heavy isotopes are distributed differently across the tag (Budnik et al., 2018; Myers et al., 2019). Thus, the fragmentation of precursor ions generates TMT reporter ions with isotopic loading unique to each TMT tag, and quantification of TMT-labeled peptides relies on reporter ions whose levels reflect both peptide abundances and noise contributions. Multiplexed peptides have an additive signal when combined, which enhances low-level samples, such as single cells.

Single Cell ProtEomics by Mass Spectrometry (SCoPE2) further exploits the additive nature of multiplexed peptides through the use of a single labeled channel that contains a significantly higher peptide concentration (Fig. 1B). This “carrier” or “boost” channel provides sufficient signal to trigger fragmentation enabling peptide sequencing, while the isobaric tags for each single cell are sufficient for obtaining relative peptide quantification. Recent work by Petelski et al. described the preparation of single mammalian macrophages, where all stages of preparation were carried out in a total volume of 1 µl (Petelski et al., 2021). Minimal sample loss and efficient sample preparation enabled over 1,000 proteins to be identified per cell.

Our group recently optimized the multiplexed single-cell proteomics methods from SCoPE2 and analyzed human-derived single-cell samples using trapped ion mobility time-of-flight mass spectrometry (Orsburn et al., 2022, preprint). When employing a peptide carrier channel to boost protein sequence coverage, we obtained over 40,000 tandem mass spectra and identified over 1,000 proteins in 30-minute LC gradients, providing unprecedented sequence coverage of each identified protein. We described the first observations of multiple classes of protein posttranslational modifications (PTMs) using liquid chromatography-mass spectrometry (LC-MS) (TIMSTOF Flex) in single human cells. PTMs play an essential role in various cellular processes that modulate the folding, stability, interaction, and functional activity of proteins (Su et al., 2017). Understanding the impact of PTMs on proteins, especially near active sites and the interfaces of protein-protein interaction, is critical for ongoing drug development research and can be leveraged to improve the efficacy of future drug design (Meng et al., 2021). Precisely targeting PTMs or protein isoforms can advance personalized treatment and precision medicine (Meng et al., 2021).

While these methods rely on traditional LC-MS, other groups have explored alternative techniques to further intensify peptide signal. Choi et al. described a custom-built capillary electrophoresis ionization platform using high-resolution mass spectrometry (Choi et al., 2021) (Fig. 1C).

This platform combines patch-clamp electrophysiology with subcellular CE-electrospray ionization-high-resolution mass spectrometry to quantify proteins in targeted neurons in brain tissues. Analysis of 1 picogram of protein digest quantified 157 proteins among the somas of three different dopaminergic neurons from a mouse (Choi et al., 2021). Additionally, Webber et al. optimized a label-free single-cell proteomics method which mitigates the slow MS2 sampling rate (Webber et al., 2022). A two-column LC system provides two parallel subsystems to multiplex sample loading, desalting, analysis, and column regeneration, which doubles the throughput as an average 621, 774, 952, and 1622 protein groups were identified with total analysis times of 7, 10, 15, and 30 minutes, respectively (Webber et al., 2022).

Small Molecule Measurements

An advancement in single-cell analysis that directly impacts pharmacology is the quantification of intracellular drug concentrations. This approach provides data regarding drug delivery, influx, and efflux at the intended target cell (i.e., CD4+ T-cells in the treatment and prevention of HIV) and can offer a real-time readout of pharmacokinetic parameters. Drug concentrations in single cells can be quantified under ambient conditions using single-probe mass spectrometry. In this technique, a probe containing dual capillaries allows for the delivery of sampling solvent and internal standard to a cell and the withdrawal of cellular extracts by capillary action. The extracts flow through a nano-electrospray ionization emitter, and ions are detected by mass spectrometry (Pan et al., 2019). Recent developments have coupled the single-probe system with an integrated cell manipulation platform, allowing for this technique to be used on cells in suspension, thereby increasing the applicability. Using this method, intracellular drug levels determined in single bladder cancer cells from patients treated with gemcitabine revealed significantly different intracellular concentrations of the drug between two patients, even after multiple rounds of treatment (Bensen et al., 2021), unveiling a need for precision medicine.

Single-probe single-cell mass spectrometry can also be used to understand the metabolic changes in cells upon treatment with drugs. These data can provide a key understanding of the drugs’ mechanisms of action and how cells can become resistant to therapeutics. This technique has been used to understand the synergism of metformin and irinotecan in an induced-irinotecan-resistant colorectal cancer cell line, where it was shown that the combination therapy had the largest synergistic effect on the reduced abundance of ceramide phosphoethanolamine, likely due to a reduction in enzymatic activity of fatty acid synthase, an enzyme overexpressed in cancer cells (Chen et al., 2022). Using this technique to identify changes in metabolite levels in single-cells upon treatment with a drug can assist in understanding the pharmacodynamics and eventual development of new combination therapies.

Another technique in development to quantify drugs in single cells is capillary electrophoresis coupled with ultrasensitive laser-induced fluorescence detection. In this method, the native fluorescence of drug molecules is exploited to quantitate single-cell drug levels. Importantly, this method can be used in tandem with flow cytometry to correlate intracellular drug levels to the abundance of cell-surface transporters, such as p-glycoprotein (Deng et al., 2011). Studies using this technique identified cell-to-cell variability in drug uptake and how dosing schedules and p-glycoprotein can impact drug uptake heterogeneity, which are factors that can have clinical implications (Deng et al., 2014).

Imaging

Finally, to decipher the molecular diversity of single cells, powerful imaging techniques, such as mass spectrometry imaging (MSI), can be used to map the localizations of a range of biomolecules including small metabolites, lipids, peptides, proteins, drugs, and drug metabolites. One of the major advantages of MSI over other imaging techniques is that it can be used to visualize the spatial distribution of hundreds of biomolecules simultaneously without labeling (Buchberger et al., 2018). The main ionization techniques used for MSI are matrix-assisted laser desorption/ionization (MALDI), desorption electrospray ionization, laser ablation electrospray ionization, and secondary ion mass spectrometry (SIMS) (Buchberger et al., 2018). Of these ionization techniques, MALDI MSI is the most frequently employed for simultaneous visualization of the spatial distribution of drugs, their metabolites, and endogenous lipid molecules at tissue level (Prideaux et al., 2010; Seneviratne et al., 2018; Tang et al., 2019; Seneviratne et al., 2020). Due to recent improvements in MALDI MS sources, this approach can also be used for single cell imaging. Several research groups have demonstrated the use of MALDI MSI to image analytes at 5 µm spatial resolution (Zavalin et al., 2013; Korte et al., 2015). However, the observed ion yields were low, and the investigators had to partially overlay individual pixels to remedy this problem (Korte et al., 2015). Technological and methodological advancements in MALDI MSI have enabled high spatial resolution; however, sensitivity is a major challenge. To overcome this, laser desorption/ionization is employed, as it avoids spectral interference due to the presence of a chemical matrix (Stopka et al., 2016). Currently, SIMS is the predominant modality for single cell MSI, and it uses a primary ion beam instead of a laser for sample ablation and ionization. Because of this, it can achieve greater spatial resolution and sensitivity than either MALDI or laser desorption/ionization. Interestingly, Malherbe et al. demonstrated that a radio frequency plasma oxygen primary ion source coupled to NanoSIMS can image endogenous cations (e.g., calcium) at a 37 nm spatial resolution (Malherbe et al., 2016). While SIMS is typically coupled to time-of-flight mass spectrometers, this ionization method can also be coupled to an orbitrap mass spectrometer to achieve higher mass resolution and improve identification of biologic molecules (Passarelli et al., 2017).

Single-cell-level imaging of endogenous metabolites can improve our understanding of the natural heterogeneity of tissues and cell types as well as enhance research in disease biology and drug discovery. These same tools can be used to study patterns of drug absorption, distribution, and metabolism. In addition to lipids and neurotransmitters, Passarelli et al. were able to detect and image the distribution of amiadarone, an antiarrhythmic drug, using OrbiSIMS (Passarelli et al., 2017). SIMS can achieve spatial resolution closer to 200 nm when imaging heavy metals and has been leveraged to image the distribution of platinum-containing drugs (Lee et al., 2017; Proetto et al., 2018).

Recently, there has been a trend toward the combined use of multiple imaging modalities (Veličković et al., 2018; Tuck et al., 2022). For example, imaging mass cytometry can obtain snapshots of tissue and cell microenvironments, and when coupled with MSI, may facilitate the correlation of drugs, drug metabolites, and endogenous molecules with cellular markers. Further, coupling microscopy and mass spectrometry can help unravel biologic processes.

Van Acker et al. demonstrated a direct comparison between fluorescence confocal microscopy and laser ablation-inductively coupled plasma-MSI techniques through the use of hybrid tracers targeting CXCR4 and EGFR (Van Acker et al., 2019). Additionally, Schoeberl et al. developed a high-throughput laser ablation-inductively coupled plasma MS workflow for single-cell analysis following cytospin preparation of cells (Schoeberl et al., 2021). Using this technique, the authors investigated cisplatin, an anticancer drug, uptake in macrophage subtypes (Schoeberl et al., 2021). The above innovative multimodal imaging strategies can be employed broadly to high-resolution imaging of drugs/metabolites and single-cell analysis in precision medicine.

In addition to these original investigations, the results of single cell studies are often available for meta-analysis through public data repositories. Today, nearly all mass spectrometry journals require that the original data and processed results are made directly accessible through the ProteomeXchange Consortium of global public repositories (Vizcaino et al., 2014). Recent efforts by the European Bioinformatics Institute repository PRIDE have enabled the public deposition of MALDI imaging data when converted to a universal XML format (Rompp et al., 2015).

With the ongoing evolution of bioinformatics tools and computational techniques, the exploration of publicly deposited single cell data will likely lead to further discoveries in addition to those made in the original studies.

Conclusion

One of the greatest obstacles in pharmacology and drug development is understanding the heterogeneity in response to drug treatment (Calabresi et al., 1979). This may arise from a number of genetic, epigenetic, and environmental factors and can be reflected by differences in morphology, physiology, and pathology (Evers et al., 2019). Intra-organ cell type heterogeneity warrants the study of distinct cell populations and cell-type-specific drug metabolism. The development of single-cell technologies will be invaluable in assessing the scope of cell-to-cell variability within a population. These advancements will provide improved analysis of an individual cell’s pharmacokinetic and pharmacodynamic responses, including expression or abundance variations in membrane drug transporters, drug metabolizing enzymes, and cellular proteomic profiling changes due to drug-induced toxicity. Further, the combination of single-cell analyses and single-cell omics can provide a wealth of knowledge and insight into biologic and pharmacological processes. We expect the elucidation of cellular heterogeneity in drug metabolism to be transformative to biomedical research and will improve our depth of understanding of biologic systems.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Wheeler, Eberhard, Mosher, Yuan, Wilkins, Seneviratne, Orsburn, Bumpus.

Footnotes

    • Received July 22, 2021.
    • Accepted October 11, 2022.
  • This work was funded by National Institutes of Health National Institute on Aging [Grant 5R01AG064908] (to N.N.B.) and National Institute of General Medical Sciences [Grant 5R01GM103853] (to N.N.B.) , National Institutes of Health [Grant T32-GM008763] (to A.M.W., C.D.E., and E.P.M) , and National Institutes of Health [Grant T32-GM144272] (to H.N.W.).

  • No author has an actual or perceived conflict of interest with the contents of this article.

  • dx.doi.org/10.1124/dmd.122.001043.

Abbreviations

CODEX
codetection by indexing
CyTOF
mass cytometry
LC-MS
liquid chromatography mass spectrometry
MALDI
matrix-assisted laser desorption/ionization
MIBI
multiplexed ion beam imaging
MSI
mass spectrometry imaging
nanoPOT
nanodroplet processing in one pot for trace sample
P450
cytochrome P450
PTM
posttranslational modification
SCoPE2
single-cell proteomics by mass spectrometry
scRNA-seq
single-cell RNA sequencing
SIMS
secondary ion mass spectrometry
SULT
sulfotransferase
TMT
tandem mass tag
UGT
UDP-glucuronosyltransferase
  • Copyright © 2023 by The Author(s)

This is an open access article distributed under the CC BY-NC Attribution 4.0 International license.

References

  1. ↵
    1. Aebersold R and
    2. Mann M
    (2003) Mass spectrometry-based proteomics. Nature 422:198–207.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Aissa AF,
    2. Islam ABMMK,
    3. Ariss MM,
    4. Go CC,
    5. Rader AE,
    6. Conrardy RD,
    7. Gajda AM,
    8. Rubio-Perez C,
    9. Valyi-Nagy K,
    10. Pasquinelli M, et al.
    (2021) Single-cell transcriptional changes associated with drug tolerance and response to combination therapies in cancer. Nat Commun 12:1628.
    OpenUrlPubMed
  3. ↵
    1. Aizarani N,
    2. Saviano A,
    3. Sagar,
    4. Mailly L,
    5. Durand S,
    6. Herman JS,
    7. Pessaux P,
    8. Baumert TF, and
    9. Grün D
    (2019) A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572:199–204.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Altschuler SJ and
    2. Wu LF
    (2010) Cellular heterogeneity: do differences make a difference? Cell 141:559–563.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Anchang B,
    2. Davis KL,
    3. Fienberg HG,
    4. Williamson BD,
    5. Bendall SC,
    6. Karacosta LG,
    7. Tibshirani R,
    8. Nolan GP, and
    9. Plevritis SK
    (2018) DRUG-NEM: Optimizing drug combinations using single-cell perturbation response to account for intratumoral heterogeneity. Proc Natl Acad Sci USA 115:E4294–E4303.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Bantscheff M,
    2. Schirle M,
    3. Sweetman G,
    4. Rick J, and
    5. Kuster B
    (2007) Quantitative mass spectrometry in proteomics: a critical review. Anal Bioanal Chem 389:1017–1031.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Barrett T,
    2. Troup DB,
    3. Wilhite SE,
    4. Ledoux P,
    5. Rudnev D,
    6. Evangelista C,
    7. Kim IF,
    8. Soboleva A,
    9. Tomashevsky M,
    10. Marshall KA, et al.
    (2009) NCBI GEO: archive for high-throughput functional genomic data. Nucleic Acids Res 37:D885–D890.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Bensen RC,
    2. Standke SJ,
    3. Colby DH,
    4. Kothapalli NR,
    5. Le-McClain AT,
    6. Patten MA,
    7. Tripathi A,
    8. Heinlen JE,
    9. Yang Z, and
    10. Burgett AWG
    (2021) Single Cell Mass Spectrometry Quantification of Anticancer Drugs: Proof of Concept in Cancer Patients. ACS Pharmacol Transl Sci 4:96–100.
    OpenUrl
  9. ↵
    1. Bloom AJ,
    2. Wang PF, and
    3. Kharasch ED
    (2019) Nicotine oxidation by genetic variants of CYP2B6 and in human brain microsomes. Pharmacol Res Perspect 7:e00468.
    OpenUrl
  10. ↵
    1. Buchberger AR,
    2. DeLaney K,
    3. Johnson J, and
    4. Li L
    (2018) Mass Spectrometry Imaging: A Review of Emerging Advancements and Future Insights. Anal Chem 90:240–265.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Budnik B,
    2. Levy E,
    3. Harmange G, and
    4. Slavov N
    (2018) SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiation. Genome Biol 19:161.
    OpenUrlCrossRef
  12. ↵
    1. Calabresi P,
    2. Dexter DL, and
    3. Heppner GH
    (1979) Clinical and pharmacological implications of cancer cell differentiation and heterogeneity. Biochem Pharmacol 28:1933–1941.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Carpenter SP,
    2. Lasker JM, and
    3. Raucy JL
    (1996) Expression, induction, and catalytic activity of the ethanol-inducible cytochrome P450 (CYP2E1) in human fetal liver and hepatocytes. Mol Pharmacol 49:260–268.
    OpenUrlAbstract
  14. ↵
    1. Carvalho AS and
    2. Matthiesen R
    (2016) Global MS-based proteomics drug profiling. Methods Mol Biol 1449:469-479.
    OpenUrl
  15. ↵
    1. Chen X,
    2. Sun M, and
    3. Yang Z
    , (2022) Single cell mass spectrometry analysis of drug-resistant cancer cells: Metabolomics studies of synergetic effect of combinational treatment. Anal Chim Acta 1201:339621.
    OpenUrl
  16. ↵
    1. Choi SB,
    2. Polter AM, and
    3. Nemes P
    (2021) Patch-Clamp Proteomics of Single Neuronal Somas in Tissue Using Electrophysiology and Subcellular Capillary Electrophoresis Mass Spectrometry. bioRxiv.
  17. ↵
    1. Coffman BL,
    2. Rios GR,
    3. King CD, and
    4. Tephly TR
    (1997) Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 25:1–4.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Courcot E,
    2. Leclerc J,
    3. Lafitte JJ,
    4. Mensier E,
    5. Jaillard S,
    6. Gosset P,
    7. Shirali P,
    8. Pottier N,
    9. Broly F, and
    10. Lo-Guidice JM
    (2012) Xenobiotic metabolism and disposition in human lung cell models: comparison with in vivo expression profiles. Drug Metab Dispos 40:1953–1965.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Court MH,
    2. Zhang X,
    3. Ding X,
    4. Yee KK,
    5. Hesse LM, and
    6. Finel M
    (2012) Quantitative distribution of mRNAs encoding the 19 human UDP-glucuronosyltransferase enzymes in 26 adult and 3 fetal tissues. Xenobiotica 42:266–277.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Ctortecka C and
    2. Mechtler K
    (2021) The rise of single‐cell proteomics. Analytical Science Advances 2:84–94.
    OpenUrl
  21. ↵
    1. Dai Y,
    2. Iwanaga K,
    3. Lin YS,
    4. Hebert MF,
    5. Davis CL,
    6. Huang W,
    7. Kharasch ED, and
    8. Thummel KE
    (2004) In vitro metabolism of cyclosporine A by human kidney CYP3A5. Biochem Pharmacol 68:1889–1902.
    OpenUrlCrossRefPubMed
  22. ↵
    1. de Graaf IA,
    2. Olinga P,
    3. de Jager MH,
    4. Merema MT,
    5. de Kanter R,
    6. van de Kerkhof EG, and
    7. Groothuis GM
    (2010) Preparation and incubation of precision-cut liver and intestinal slices for application in drug metabolism and toxicity studies. Nat Protoc 5:1540–1551.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Deng B,
    2. Wang Z,
    3. Song J,
    4. Xiao Y,
    5. Chen D, and
    6. Huang J
    (2011) Analysis of doxorubicin uptake in single human leukemia K562 cells using capillary electrophoresis coupled with laser-induced fluorescence detection. Anal Bioanal Chem 401:2143–2152.
    OpenUrlPubMed
  24. ↵
    1. Deng B,
    2. Wang ZM,
    3. Zhou ZH,
    4. Liu YM,
    5. Yang XL,
    6. Song J, and
    7. Xiao YX
    (2014) Quantitative study of cellular heterogeneity in doxorubicin uptake and its pharmacological effect on cancer cells. Biomed Chromatogr 28:1393–1401.
    OpenUrl
  25. ↵
    1. Evers TM,
    2. Hochane M,
    3. Tans SJ,
    4. Heeren RM,
    5. Semrau S,
    6. Nemes P, and
    7. Mashaghi A
    (2019) Deciphering metabolic heterogeneity by single-cell analysis, ACS Publications.
  26. ↵
    1. Fonsi M
    (2014) Extrahepatic metabolism may complicate the IVIVC in rats. Drug Metab Lett 8:51–66.
    OpenUrl
  27. ↵
    1. Gay L,
    2. Baker A-M, and
    3. Graham TA
    (2016) Tumour Cell Heterogeneity. F1000 Res 5:F1000.
    OpenUrl
  28. ↵
    1. Ghosh C,
    2. Gonzalez-Martinez J,
    3. Hossain M,
    4. Cucullo L,
    5. Fazio V,
    6. Janigro D, and
    7. Marchi N
    (2010) Pattern of P450 expression at the human blood-brain barrier: roles of epileptic condition and laminar flow. Epilepsia 51:1408–1417.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Glasser MF,
    2. Coalson TS,
    3. Robinson EC,
    4. Hacker CD,
    5. Harwell J,
    6. Yacoub E,
    7. Ugurbil K,
    8. Andersson J,
    9. Beckmann CF,
    10. Jenkinson M, et al.
    (2016) A multi-modal parcellation of human cerebral cortex. Nature 536:171–178.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Goltsev Y,
    2. Samusik N,
    3. Kennedy-Darling J,
    4. Bhate S,
    5. Hale M,
    6. Vazquez G,
    7. Black S, and
    8. Nolan GP
    (2018) Deep Profiling of Mouse Splenic Architecture with CODEX Multiplexed Imaging. Cell 174:968–981.e15.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Guan J,
    2. Lin Y,
    3. Wang Y,
    4. Gao J, and
    5. Ji G
    (2021) An analytical method for the identification of cell type-specific disease gene modules. J Transl Med 19:20.
    OpenUrl
  32. ↵
    1. Gundry RL,
    2. White MY,
    3. Murray CI,
    4. Kane LA,
    5. Fu Q,
    6. Stanley BA, and
    7. Van Eyk JE
    (2009) Preparation of proteins and peptides for mass spectrometry analysis in a bottom‐up proteomics workflow. Curr Protoc Mol Biol 90:10.25. 11-10.25.23.
  33. ↵
    1. Hartlmayr D,
    2. Ctortecka C,
    3. Seth A,
    4. Mendjan S,
    5. Tourniaire G, and
    6. Mechtler K
    (2021) An automated workflow for label-free and multiplexed single cell proteomics sample preparation at unprecedented sensitivity. bioRxiv.
  34. ↵
    1. Hiraoka H,
    2. Yamamoto K,
    3. Miyoshi S,
    4. Morita T,
    5. Nakamura K,
    6. Kadoi Y,
    7. Kunimoto F, and
    8. Horiuchi R
    (2005) Kidneys contribute to the extrahepatic clearance of propofol in humans, but not lungs and brain. Br J Clin Pharmacol 60:176–182.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Ho MD,
    2. Ring N,
    3. Amaral K,
    4. Doshi U, and
    5. Li AP
    (2017) Human Enterocytes as an In Vitro Model for the Evaluation of Intestinal Drug Metabolism: Characterization of Drug-Metabolizing Enzyme Activities of Cryopreserved Human Enterocytes from Twenty-Four Donors. Drug Metab Dispos 45:686–691.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Hodge RD,
    2. Bakken TE,
    3. Miller JA,
    4. Smith KA,
    5. Barkan ER,
    6. Graybuck LT,
    7. Close JL,
    8. Long B,
    9. Johansen N,
    10. Penn O, et al.
    (2019) Conserved cell types with divergent features in human versus mouse cortex. Nature 573:61–68.
    OpenUrlCrossRef
  37. ↵
    1. Iswandana R,
    2. Irianti MI,
    3. Oosterhuis D,
    4. Hofker HS,
    5. Merema MT,
    6. de Jager MH,
    7. Mutsaers HAM, and
    8. Olinga P
    (2018) Regional Differences in Human Intestinal Drug Metabolism. Drug Metab Dispos 46:1879–1885.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Izar B,
    2. Tirosh I,
    3. Stover EH,
    4. Wakiro I,
    5. Cuoco MS,
    6. Alter I,
    7. Rodman C,
    8. Leeson R,
    9. Su M-J,
    10. Shah P, et al.
    (2020) A single-cell landscape of high-grade serous ovarian cancer. Nat Med 26:1271–1279.
    OpenUrlPubMed
  39. ↵
    1. Jones NR and
    2. Lazarus P
    (2014) UGT2B gene expression analysis in multiple tobacco carcinogen-targeted tissues. Drug Metab Dispos 42:529–536.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    1. Kaut O,
    2. Schmitt I, and
    3. Wüllner U
    (2012) Genome-scale methylation analysis of Parkinson’s disease patients’ brains reveals DNA hypomethylation and increased mRNA expression of cytochrome P450 2E1. Neurogenetics 13:87–91.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Kelly R,
    2. Zhu Y,
    3. Liang Y,
    4. Cong Y,
    5. Piehowski P,
    6. Dou M,
    7. Zhao R,
    8. Qian W-J,
    9. Burnum-Johnson K, and
    10. Ansong C
    (2019) Single Cell Proteome Mapping of Tissue Heterogeneity Using Microfluidic Nanodroplet Sample Processing and Ultrasensitive LC-MS. Journal of Biomolecular Techniques: JBT 30:S61.
    OpenUrl
  42. ↵
    1. Keren L,
    2. Bosse M,
    3. Thompson S,
    4. Risom T,
    5. Vijayaragavan K,
    6. McCaffrey E,
    7. Marquez D,
    8. Angoshtari R,
    9. Greenwald NF,
    10. Fienberg H, et al.
    (2019) MIBI-TOF: A multiplexed imaging platform relates cellular phenotypes and tissue structure. Sci Adv 5:eaax5851.
    OpenUrlFREE Full Text
  43. ↵
    1. Kim JH,
    2. Sherman ME,
    3. Curriero FC,
    4. Guengerich FP,
    5. Strickland PT, and
    6. Sutter TR
    (2004) Expression of cytochromes P450 1A1 and 1B1 in human lung from smokers, non-smokers, and ex-smokers. Toxicol Appl Pharmacol 199:210–219.
    OpenUrlCrossRefPubMed
  44. ↵
    1. King CD,
    2. Rios GR,
    3. Assouline JA, and
    4. Tephly TR
    (1999) Expression of UDP-glucuronosyltransferases (UGTs) 2B7 and 1A6 in the human brain and identification of 5-hydroxytryptamine as a substrate. Arch Biochem Biophys 365:156–162.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Kirschner K,
    2. Rattanavirotkul N,
    3. Quince MF, and
    4. Chandra T
    (2020) Functional heterogeneity in senescence. Biochem Soc Trans 48:765–773.
    OpenUrlCrossRef
  46. ↵
    1. Knights KM,
    2. Spencer SM,
    3. Fallon JK,
    4. Chau N,
    5. Smith PC, and
    6. Miners JO
    (2016) Scaling factors for the in vitro-in vivo extrapolation (IV-IVE) of renal drug and xenobiotic glucuronidation clearance. Br J Clin Pharmacol 81:1153–1164.
    OpenUrl
  47. ↵
    1. Kolars JC,
    2. Awni WM,
    3. Merion RM, and
    4. Watkins PB
    (1991) First-pass metabolism of cyclosporin by the gut. Lancet 338:1488–1490.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Korte AR,
    2. Yandeau-Nelson MD,
    3. Nikolau BJ, and
    4. Lee YJ
    (2015) Subcellular-level resolution MALDI-MS imaging of maize leaf metabolites by MALDI-linear ion trap-Orbitrap mass spectrometer. Anal Bioanal Chem 407:2301–2309.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Kuban W and
    2. Daniel WA
    (2021) Cytochrome P450 expression and regulation in the brain. Drug Metab Rev 53:1–29.
    OpenUrl
  50. ↵
    1. Lash LH,
    2. Putt DA, and
    3. Cai H
    (2008) Drug metabolism enzyme expression and activity in primary cultures of human proximal tubular cells. Toxicology 244:56–65.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Lee RFS,
    2. Theiner S,
    3. Meibom A,
    4. Koellensperger G,
    5. Keppler BK, and
    6. Dyson PJ
    (2017) Application of imaging mass spectrometry approaches to facilitate metal-based anticancer drug research. Metallomics 9:365–381.
    OpenUrl
  52. ↵
    1. Li AP
    (2020) In Vitro Human Cell-Based Experimental Models for the Evaluation of Enteric Metabolism and Drug Interaction Potential of Drugs and Natural Products. Drug Metab Dispos 48:980–992.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    1. Liao J,
    2. Yu Z,
    3. Chen Y,
    4. Bao M,
    5. Zou C,
    6. Zhang H,
    7. Liu D,
    8. Li T,
    9. Zhang Q,
    10. Li J, et al.
    (2020) Single-cell RNA sequencing of human kidney. Sci Data 7:4.
    OpenUrl
  54. ↵
    1. Lin D,
    2. Chen Y,
    3. Negi S,
    4. Cheng D,
    5. Ouyang Z,
    6. Sexton D,
    7. Li K, and
    8. Zhang B
    (2022) CellDepot: A Unified Repository for scRNA-seq Data and Visual Exploration. J Mol Biol 434:167425.
    OpenUrl
  55. ↵
    1. Macosko EZ,
    2. Basu A,
    3. Satija R,
    4. Nemesh J,
    5. Shekhar K,
    6. Goldman M,
    7. Tirosh I,
    8. Bialas AR,
    9. Kamitaki N,
    10. Martersteck EM, et al.
    (2015) Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell 161:1202–1214.
    OpenUrlCrossRefPubMed
  56. ↵
    1. MacParland SA,
    2. Liu JC,
    3. Ma XZ,
    4. Innes BT,
    5. Bartczak AM,
    6. Gage BK,
    7. Manuel J,
    8. Khuu N,
    9. Echeverri J,
    10. Linares I, et al.
    (2018) Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat Commun 9:4383.
    OpenUrlCrossRefPubMed
  57. ↵
    1. Malherbe J,
    2. Penen F,
    3. Isaure MP,
    4. Frank J,
    5. Hause G,
    6. Dobritzsch D,
    7. Gontier E,
    8. Horréard F,
    9. Hillion F, and
    10. Schaumlöffel D
    (2016) A New Radio Frequency Plasma Oxygen Primary Ion Source on Nano Secondary Ion Mass Spectrometry for Improved Lateral Resolution and Detection of Electropositive Elements at Single Cell Level. Anal Chem 88:7130–7136.
    OpenUrl
  58. ↵
    1. Marcu R,
    2. Choi YJ,
    3. Xue J,
    4. Fortin CL,
    5. Wang Y,
    6. Nagao RJ,
    7. Xu J,
    8. MacDonald JW,
    9. Bammler TK,
    10. Murry CE, et al.
    (2018) Human Organ-Specific Endothelial Cell Heterogeneity. iScience 4:20–35.
    OpenUrl
  59. ↵
    1. Marx V
    (2019) A dream of single-cell proteomics. Nat Methods 16:809–812.
    OpenUrlCrossRef
  60. ↵
    1. McFadyen MCE,
    2. Melvin WT, and
    3. Murray GI
    (1998) Regional distribution of individual forms of cytochrome P450 mRNA in normal adult human brain. Biochem Pharmacol 55:825–830.
    OpenUrlCrossRefPubMed
  61. ↵
    1. McGinnity DF,
    2. Soars MG,
    3. Urbanowicz RA, and
    4. Riley RJ
    (2004) Evaluation of fresh and cryopreserved hepatocytes as in vitro drug metabolism tools for the prediction of metabolic clearance. Drug Metab Dispos 32:1247–1253.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Meissner F,
    2. Geddes-McAlister J,
    3. Mann M, and
    4. Bantscheff M
    (2022) The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov 21:637–654.
    OpenUrl
  63. ↵
    1. Meng F,
    2. Liang Z,
    3. Zhao K, and
    4. Luo C
    (2021) Drug design targeting active posttranslational modification protein isoforms. Med Res Rev 41:1701–1750.
    OpenUrl
  64. ↵
    1. Moreno P,
    2. Fexova S,
    3. George N,
    4. Manning JR,
    5. Miao Z,
    6. Mohammed S,
    7. Muñoz-Pomer A,
    8. Fullgrabe A,
    9. Bi Y,
    10. Bush N, et al.
    (2022) Expression Atlas update: gene and protein expression in multiple species. Nucleic Acids Res 50 (D1):D129–D140.
    OpenUrl
  65. ↵
    1. Myers SA,
    2. Klaeger S,
    3. Satpathy S,
    4. Viner R,
    5. Choi J,
    6. Rogers J,
    7. Clauser K,
    8. Udeshi ND, and
    9. Carr SA
    (2019) Evaluation of Advanced Precursor Determination for Tandem Mass Tag (TMT)-Based Quantitative Proteomics across Instrument Platforms. J Proteome Res 18:542–547.
    OpenUrl
  66. ↵
    1. Norton SE,
    2. Ward-Hartstonge KA,
    3. McCall JL,
    4. Leman JKH,
    5. Taylor ES,
    6. Munro F,
    7. Black MA,
    8. Fazekas de St Groth B,
    9. McGuire HM, and
    10. Kemp RA
    (2019) High-Dimensional Mass Cytometric Analysis Reveals an Increase in Effector Regulatory T Cells as a Distinguishing Feature of Colorectal Tumors. J Immunol 202:1871–1884.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Olsen JV and
    2. Mann M
    (2013) Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol Cell Proteomics 12:3444–3452.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Paine MF,
    2. Shen DD,
    3. Kunze KL,
    4. Perkins JD,
    5. Marsh CL,
    6. McVicar JP,
    7. Barr DM,
    8. Gillies BS, and
    9. Thummel KE
    (1996) First-pass metabolism of midazolam by the human intestine. Clin Pharmacol Ther 60:14–24.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Pan N,
    2. Standke SJ,
    3. Kothapalli NR,
    4. Sun M,
    5. Bensen RC,
    6. Burgett AWG, and
    7. Yang Z
    (2019) Quantification of Drug Molecules in Live Single Cells Using the Single-Probe Mass Spectrometry Technique. Anal Chem 91:9018–9024.
    OpenUrl
  70. ↵
    1. Passarelli MK,
    2. Pirkl A,
    3. Moellers R,
    4. Grinfeld D,
    5. Kollmer F,
    6. Havelund R,
    7. Newman CF,
    8. Marshall PS,
    9. Arlinghaus H,
    10. Alexander MR, et al.
    (2017) The 3D OrbiSIMS-label-free metabolic imaging with subcellular lateral resolution and high mass-resolving power. Nat Methods 14:1175–1183.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Paszek P,
    2. Ryan S,
    3. Ashall L,
    4. Sillitoe K,
    5. Harper CV,
    6. Spiller DG,
    7. Rand DA, and
    8. White MR
    (2010) Population robustness arising from cellular heterogeneity. Proc Natl Acad Sci USA 107:11644–11649.
    OpenUrlAbstract/FREE Full Text
  72. ↵
    1. Payen VL,
    2. Lavergne A,
    3. Alevra Sarika N,
    4. Colonval M,
    5. Karim L,
    6. Deckers M,
    7. Najimi M,
    8. Coppieters W,
    9. Charloteaux B,
    10. Sokal EM, et al.
    (2021) Single-cell RNA sequencing of human liver reveals hepatic stellate cell heterogeneity. JHEP Rep 3:100278.
    OpenUrlCrossRef
  73. ↵
    1. Peng J,
    2. Sun B-F,
    3. Chen C-Y,
    4. Zhou J-Y,
    5. Chen Y-S,
    6. Chen H,
    7. Liu L,
    8. Huang D,
    9. Jiang J,
    10. Cui G-S, et al.
    (2019) Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res 29:725–738.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Petelski AA,
    2. Emmott E,
    3. Leduc A,
    4. Huffman RG,
    5. Specht H,
    6. Perlman DH, and
    7. Slavov N
    (2021) Multiplexed single-cell proteomics using SCoPE2. Nat Protoc 16:5398–5425.
    OpenUrl
  75. ↵
    1. Ponsoda X,
    2. Pareja E,
    3. Gómez-Lechón MJ,
    4. Fabra R,
    5. Carrasco E,
    6. Trullenque R, and
    7. Castell JV
    (2001) Drug biotransformation by human hepatocytes. In vitro/in vivo metabolism by cells from the same donor. J Hepatol 34:19–25.
    OpenUrlPubMed
  76. ↵
    1. Prideaux B,
    2. Staab D, and
    3. Stoeckli M
    (2010) Applications of MALDI-MSI to pharmaceutical research. Methods Mol Biol 656:405–413.
    OpenUrlCrossRefPubMed
  77. ↵
    1. Proetto MT,
    2. Callmann CE,
    3. Cliff J,
    4. Szymanski CJ,
    5. Hu D,
    6. Howell SB,
    7. Evans JE,
    8. Orr G, and
    9. Gianneschi NC
    (2018) Tumor Retention of Enzyme-Responsive Pt(II) Drug-Loaded Nanoparticles Imaged by Nanoscale Secondary Ion Mass Spectrometry and Fluorescence Microscopy. ACS Cent Sci 4:1477–1484.
    OpenUrl
  78. ↵
    1. Riches Z,
    2. Stanley EL,
    3. Bloomer JC, and
    4. Coughtrie MW
    (2009) Quantitative evaluation of the expression and activity of five major sulfotransferases (SULTs) in human tissues: the SULT “pie”. Drug Metab Dispos 37:2255–2261.
    OpenUrlAbstract/FREE Full Text
  79. ↵
    1. Rinke C,
    2. Lee J,
    3. Nath N,
    4. Goudeau D,
    5. Thompson B,
    6. Poulton N,
    7. Dmitrieff E,
    8. Malmstrom R,
    9. Stepanauskas R, and
    10. Woyke T
    (2014) Obtaining genomes from uncultivated environmental microorganisms using FACS-based single-cell genomics. Nat Protoc 9:1038–1048.
    OpenUrlCrossRefPubMed
    1. Römpp A,
    2. Both JP,
    3. Brunelle A,
    4. Heeren RM,
    5. Laprévote O,
    6. Prideaux B,
    7. Seyer A,
    8. Spengler B,
    9. Stoeckli M, and
    10. Smith DF
    (2015) Mass spectrometry imaging of biological tissue: an approach for multicenter studies. Anal Bioanal Chem 407:2329–2335.
    OpenUrl
  80. ↵
    1. Salman ED,
    2. Kadlubar SA, and
    3. Falany CN
    (2009) Expression and localization of cytosolic sulfotransferase (SULT) 1A1 and SULT1A3 in normal human brain. Drug Metab Dispos 37:706–709.
    OpenUrlAbstract/FREE Full Text
  81. ↵
    1. Schoeberl A,
    2. Gutmann M,
    3. Theiner S,
    4. Schaier M,
    5. Schweikert A,
    6. Berger W, and
    7. Koellensperger G
    (2021) Cisplatin Uptake in Macrophage Subtypes at the Single-Cell Level by LA-ICP-TOFMS Imaging. Anal Chem 93:16456–16465.
    OpenUrl
  82. ↵
    1. Seneviratne HK,
    2. Hamlin AN,
    3. Heck CJS, and
    4. Bumpus NN
    (2020) Spatial Distribution Profiles of Emtricitabine, Tenofovir, Efavirenz, and Rilpivirine in Murine Tissues Following In Vivo Dosing Correlate with Their Safety Profiles in Humans. ACS Pharmacol Transl Sci 3:655–665.
    OpenUrl
  83. ↵
    1. Seneviratne HK,
    2. Hendrix CW,
    3. Fuchs EJ, and
    4. Bumpus NN
    (2018) MALDI Mass Spectrometry Imaging Reveals Heterogeneous Distribution of Tenofovir and Tenofovir Diphosphate in Colorectal Tissue of Subjects Receiving a Tenofovir-Containing Enema. J Pharmacol Exp Ther 367:40–48.
    OpenUrlAbstract/FREE Full Text
  84. ↵
    1. Sergent T,
    2. Dupont I,
    3. Van der Heiden E,
    4. Scippo ML,
    5. Pussemier L,
    6. Larondelle Y, and
    7. Schneider YJ
    (2009) CYP1A1 and CYP3A4 modulation by dietary flavonoids in human intestinal Caco-2 cells. Toxicol Lett 191:216–222.
    OpenUrlCrossRefPubMed
  85. ↵
    1. Shahabi HN,
    2. Andersson DR, and
    3. Nissbrandt H
    (2008) Cytochrome P450 2E1 in the substantia nigra: relevance for dopaminergic neurotransmission and free radical production. Synapse 62:379–388.
    OpenUrlCrossRefPubMed
  86. ↵
    1. Shawahna R,
    2. Uchida Y,
    3. Declèves X,
    4. Ohtsuki S,
    5. Yousif S,
    6. Dauchy S,
    7. Jacob A,
    8. Chassoux F,
    9. Daumas-Duport C,
    10. Couraud PO, et al.
    (2011) Transcriptomic and quantitative proteomic analysis of transporters and drug metabolizing enzymes in freshly isolated human brain microvessels. Mol Pharm 8:1332–1341.
    OpenUrlCrossRefPubMed
  87. ↵
    1. Shibata Y and
    2. Chiba M
    (2015) The role of extrahepatic metabolism in the pharmacokinetics of the targeted covalent inhibitors afatinib, ibrutinib, and neratinib. Drug Metab Dispos 43:375–384.
    OpenUrlAbstract/FREE Full Text
  88. ↵
    1. Slavov N
    (2022) Scaling Up Single-Cell Proteomics. Mol Cell Proteomics 21:100179.
    OpenUrl
  89. ↵
    1. Somers GI,
    2. Lindsay N,
    3. Lowdon BM,
    4. Jones AE,
    5. Freathy C,
    6. Ho S,
    7. Woodrooffe AJ,
    8. Bayliss MK, and
    9. Manchee GR
    (2007) A comparison of the expression and metabolizing activities of phase I and II enzymes in freshly isolated human lung parenchymal cells and cryopreserved human hepatocytes. Drug Metab Dispos 35:1797–1805.
    OpenUrlAbstract/FREE Full Text
  90. ↵
    1. Stopka SA,
    2. Rong C,
    3. Korte AR,
    4. Yadavilli S,
    5. Nazarian J,
    6. Razunguzwa TT,
    7. Morris NJ, and
    8. Vertes A
    (2016) Molecular Imaging of Biological Samples on Nanophotonic Laser Desorption Ionization Platforms. Angew Chem Int Ed Engl 55:4482–4486.
    OpenUrl
  91. ↵
    1. Su M-G,
    2. Weng JT-Y,
    3. Hsu JB-K,
    4. Huang K-Y,
    5. Chi Y-H, and
    6. Lee T-Y
    (2017) Investigation and identification of functional post-translational modification sites associated with drug binding and protein-protein interactions. BMC Syst Biol 11 (Suppl 7):132.
    OpenUrl
  92. ↵
    1. Takayama K,
    2. Ito K,
    3. Matsui A,
    4. Yamashita T,
    5. Kawakami K,
    6. Hirayama D,
    7. Kishimoto W,
    8. Nakase H, and
    9. Mizuguchi H
    (2021) In Vivo Gene Expression Profile of Human Intestinal Epithelial Cells: From the Viewpoint of Drug Metabolism and Pharmacokinetics. Drug Metab Dispos 49:221–232.
    OpenUrlAbstract/FREE Full Text
  93. ↵
    1. Tang W,
    2. Chen J,
    3. Zhou J,
    4. Ge J,
    5. Zhang Y,
    6. Li P, and
    7. Li B
    (2019) Quantitative MALDI Imaging of Spatial Distributions and Dynamic Changes of Tetrandrine in Multiple Organs of Rats. Theranostics 9:932–944.
    OpenUrl
  94. ↵
    1. Thummel KE,
    2. O’Shea D,
    3. Paine MF,
    4. Shen DD,
    5. Kunze KL,
    6. Perkins JD, and
    7. Wilkinson GR
    (1996) Oral first-pass elimination of midazolam involves both gastrointestinal and hepatic CYP3A-mediated metabolism. Clin Pharmacol Ther 59:491–502.
    OpenUrlCrossRefPubMed
  95. ↵
    1. Toselli F,
    2. Booth Depaz IM,
    3. Worrall S,
    4. Etheridge N,
    5. Dodd PR,
    6. Wilce PA, and
    7. Gillam EM
    (2015) Expression of CYP2E1 and CYP2U1 proteins in amygdala and prefrontal cortex: influence of alcoholism and smoking. Alcohol Clin Exp Res 39:790–797.
    OpenUrl
  96. ↵
    1. Tracey LJ,
    2. An Y, and
    3. Justice MJ
    (2021) CyTOF: An Emerging Technology for Single-Cell Proteomics in the Mouse. Curr Protoc 1:e118.
    OpenUrl
  97. ↵
    1. Travaglini KJ,
    2. Nabhan AN,
    3. Penland L,
    4. Sinha R,
    5. Gillich A,
    6. Sit RV,
    7. Chang S,
    8. Conley SD,
    9. Mori Y,
    10. Seita J, et al.
    (2020) A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587:619–625.
    OpenUrlCrossRefPubMed
  98. ↵
    1. Tuck M,
    2. Grélard F,
    3. Blanc L, and
    4. Desbenoit N
    (2022) MALDI-MSI Towards Multimodal Imaging: Challenges and Perspectives. Front Chem 10:904688.
    OpenUrl
  99. ↵
    1. Upadhya SC,
    2. Tirumalai PS,
    3. Boyd MR,
    4. Mori T, and
    5. Ravindranath V
    (2000) Cytochrome P4502E (CYP2E) in brain: constitutive expression, induction by ethanol and localization by fluorescence in situ hybridization. Arch Biochem Biophys 373:23–34.
    OpenUrlCrossRefPubMed
  100. ↵
    1. Van Acker T,
    2. Buckle T,
    3. Van Malderen SJM,
    4. van Willigen DM,
    5. van Unen V,
    6. van Leeuwen FWB, and
    7. Vanhaecke F
    (2019) High-resolution imaging and single-cell analysis via laser ablation-inductively coupled plasma-mass spectrometry for the determination of membranous receptor expression levels in breast cancer cell lines using receptor-specific hybrid tracers. Anal Chim Acta 1074:43–53.
    OpenUrl
  101. ↵
    1. Van der Hauwaert C,
    2. Savary G,
    3. Buob D,
    4. Leroy X,
    5. Aubert S,
    6. Flamand V,
    7. Hennino MF,
    8. Perrais M,
    9. Lo-Guidice JM,
    10. Broly F, et al.
    (2014) Expression profiles of genes involved in xenobiotic metabolism and disposition in human renal tissues and renal cell models. Toxicol Appl Pharmacol 279:409–418.
    OpenUrl
  102. ↵
    1. van Herwaarden AE,
    2. Wagenaar E,
    3. van der Kruijssen CM,
    4. van Waterschoot RA,
    5. Smit JW,
    6. Song JY,
    7. van der Valk MA,
    8. van Tellingen O,
    9. van der Hoorn JW,
    10. Rosing H, et al.
    (2007) Knockout of cytochrome P450 3A yields new mouse models for understanding xenobiotic metabolism. J Clin Invest 117:3583–3592.
    OpenUrlCrossRefPubMed
  103. ↵
    1. Veličković D,
    2. Chu RK,
    3. Carrell AA,
    4. Thomas M,
    5. Paša-Tolić L,
    6. Weston DJ, and
    7. Anderton CR
    , (2018) Multimodal MSI in Conjunction with Broad Coverage Spatially Resolved MS2 Increases Confidence in Both Molecular Identification and Localization. Anal Chem 90:702–707.
    OpenUrl
    1. Vizcaíno JA,
    2. Deutsch EW,
    3. Wang R,
    4. Csordas A,
    5. Reisinger F,
    6. Ríos D,
    7. Dianes JA,
    8. Sun Z,
    9. Farrah T,
    10. Bandeira N, et al.
    (2014) ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat Biotechnol 32:223–226.
    OpenUrlCrossRefPubMed
    1. Wahlström A,
    2. Winblad B,
    3. Bixo M, and
    4. Rane A
    (1988) Human brain metabolism of morphine and naloxone. Pain 35:121–127.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Webber KG,
    2. Truong T,
    3. Johnston SM,
    4. Zapata SE,
    5. Liang Y,
    6. Davis JM,
    7. Buttars AD,
    8. Smith FB,
    9. Jones HE, and
    10. Mahoney AC
    (2022) Label-Free Profiling of up to 200 Single-Cell Proteomes per Day Using a Dual-Column Nanoflow Liquid Chromatography Platform. Anal Chem 15:6017–6025.
    OpenUrl
  105. ↵
    1. Wilting RH and
    2. Dannenberg J-H
    (2012) Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance. Drug Resist Updat 15:21–38.
    OpenUrlCrossRefPubMed
  106. ↵
    1. Xu P,
    2. Wang M,
    3. Song WM,
    4. Wang Q,
    5. Yuan GC,
    6. Sudmant PH,
    7. Zare H,
    8. Tu Z,
    9. Orr ME, and
    10. Zhang B
    (2022) The landscape of human tissue and cell type specific expression and co-regulation of senescence genes. Mol Neurodegener 17:5.
    OpenUrl
  107. ↵
    1. Zavalin A,
    2. Yang J, and
    3. Caprioli R
    (2013) Laser beam filtration for high spatial resolution MALDI imaging mass spectrometry. J Am Soc Mass Spectrom 24:1153–1156.
    OpenUrl
  108. ↵
    1. Zhang Q,
    2. Lou Y,
    3. Yang J,
    4. Wang J,
    5. Feng J,
    6. Zhao Y,
    7. Wang L,
    8. Huang X,
    9. Fu Q,
    10. Ye M, et al.
    (2019) Integrated multiomic analysis reveals comprehensive tumour heterogeneity and novel immunophenotypic classification in hepatocellular carcinomas. Gut 68:2019–2031.
    OpenUrlAbstract/FREE Full Text
  109. ↵
    1. Zhang QY,
    2. Kaminsky LS,
    3. Dunbar D,
    4. Zhang J, and
    5. Ding X
    (2007) Role of small intestinal cytochromes p450 in the bioavailability of oral nifedipine. Drug Metab Dispos 35:1617–1623.
    OpenUrlAbstract/FREE Full Text
  110. ↵
    1. Zhong Y,
    2. Dong G,
    3. Luo H,
    4. Cao J,
    5. Wang C,
    6. Wu J,
    7. Feng YQ, and
    8. Yue J
    (2012) Induction of brain CYP2E1 by chronic ethanol treatment and related oxidative stress in hippocampus, cerebellum, and brainstem. Toxicology 302:275–284.
    OpenUrl
  111. ↵
    1. Zhu K,
    2. Liu X,
    3. Liu C,
    4. Xu Y,
    5. Fu Y,
    6. Dong W,
    7. Yan Y,
    8. Wang W, and
    9. Qian C
    (2021) AKT inhibitor AZD5363 suppresses stemness and promotes anti-cancer activity of 3,3′-diindolylmethane in human breast cancer cells. Toxicol Appl Pharmacol 429:115700.
    OpenUrl
  112. ↵
    1. Zhu Y,
    2. Piehowski PD,
    3. Zhao R,
    4. Chen J,
    5. Shen Y,
    6. Moore RJ,
    7. Shukla AK,
    8. Petyuk VA,
    9. Campbell-Thompson M,
    10. Mathews CE, et al.
    (2018) Nanodroplet processing platform for deep and quantitative proteome profiling of 10-100 mammalian cells. Nat Commun 9:882.
    OpenUrlCrossRefPubMed
  113. ↵
    1. Zimatkin SM,
    2. Pronko SP,
    3. Vasiliou V,
    4. Gonzalez FJ, and
    5. Deitrich RA
    (2006) Enzymatic mechanisms of ethanol oxidation in the brain. Alcohol Clin Exp Res 30:1500–1505.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 51 (3)
Drug Metabolism and Disposition
Vol. 51, Issue 3
1 Mar 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Achieving a Deeper Understanding of Drug Metabolism and Responses Using Single-Cell Technologies
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Review Article50th Anniversary Celebration Collection Special Section on Drug Metabolism and Precision Medicine—Minireview

Single Cell Technologies for Pharmacology

Abigail M. Wheeler, Colten D. Eberhard, Eric P. Mosher, Yuting Yuan, Hannah N. Wilkins, Herana Kamal Seneviratne, Benjamin C. Orsburn and Namandjé N. Bumpus
Drug Metabolism and Disposition March 1, 2023, 51 (3) 350-359; DOI: https://doi.org/10.1124/dmd.122.001043

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Review Article50th Anniversary Celebration Collection Special Section on Drug Metabolism and Precision Medicine—Minireview

Single Cell Technologies for Pharmacology

Abigail M. Wheeler, Colten D. Eberhard, Eric P. Mosher, Yuting Yuan, Hannah N. Wilkins, Herana Kamal Seneviratne, Benjamin C. Orsburn and Namandjé N. Bumpus
Drug Metabolism and Disposition March 1, 2023, 51 (3) 350-359; DOI: https://doi.org/10.1124/dmd.122.001043
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Conclusion
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Structure–Function Relationship of the CYP2B6 Active Site
  • Tissue specific metabolism of nucleoside/nucleotide drugs
Show more 50th Anniversary Celebration Collection Special Section on Drug Metabolism and Precision Medicine—Minireview

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics