Cell Proliferation Assays and Cell Viability Assays
Laura Cobb (laura at lcsexecutiveconsulting dot com)
University of California at Los Angeles, United States (till January, 2012); LCS Executive Consulting, Los Angeles, United States
DOI
//dx.doi.org/10.13070/mm.en.3.2799
Date
last modified : 2023-12-25; original version : 2013-03-05
Cite as
MATER METHODS 2013;3:2799
Abstract

A comprehensive review of assays for cell proliferation and cell viability, along with the results from a Labome survey of formal publications.

Cell Proliferation Assays

There are many methods and kits currently available that are used to measure numerous aspects of cell proliferation, mitochondrial function and, indirectly, cell viability. However, over-analysis of these assays and incorrect interpretation of the information they provide are becoming more frequent in the published literature. In a cell proliferation assay, the output should give you a direct and accurate measurement of the number of actively dividing cells in a population, be it cells in culture or tissues. In contrast, a cell viability assay is designed to provide an indication of the number of “healthy” cells within a population, frequently by assessing specific indicators of metabolically active cells, often related to mitochondrial function. Unlike a proliferation assay, these measurements do not distinguish between quiescent/senescent and actively dividing cells. Beta-galactosidase activity assay or staining can indicate senescent cells [1]. Assays for the cell cycle analysis and cell death are discussed elsewhere.

Label-free approaches, such as xCELLigence RTCA SP/DP Real Time Cell Analyzer from ACEA Biosciences, can measure the increase in cell numbers, and thus quantify the cell adhesion, proliferation, detachment/death [2, 3].

Cell Proliferation Assays and Cell Viability Assays figure 1
Figure 1. Method for measuring BrdU incorporation
Assessing DNA synthesis

The traditional method for assaying cell proliferation is to measure DNA synthesis by assessing the incorporation of a labeled DNA analog or precursor (5-bromo-2’-deoxyuridine (BrdU [4] ), an analog of pyrimidine which gets incorporated it to new DNA in the place of thymidine, or [3H]-thymidine) into the genomic DNA of cells during S phase of the cell cycle. For example, Maun HR et al assessed the proliferation of bronchial smooth muscle cells with 3H-thymidine incorporation [5]. For cells in culture, the most common method is to assess BrdU incorporation by colorimetric ELISA (Figure 1). Similar methods can be used to assess proliferating cells in vivo by pulse-labeling tissues with BrdU before harvesting, followed by assessing BrdU by ELISA or immunohistochemical staining. This can also be assessed by flow cytometry; an example trace of BrdU vs. 7-AAD is shown in Figure 2.

A newer approach is to incorporate the alkyne-containing thymidine analog EdU (5-ethynyl-2´-deoxyuridine) into DNA and detect the incorporated EdU by a click reaction—a copper-catalyzed azide–alkyne cycloaddition [6] - in the case of labeling in live animals, EdU can be either injected or mixed in the drinking water [7]. Ouadah Y et al injected, i.p., BrdU and Edu daily into mice and detected the labelled cells through an anti-BrdU antibody (Abcam ab6326) and Click-iT EdU kits from Thermo Fisher on the cryosections to investigate the activation of pulmonary neuroendocrine cells [8]. A Marconi et al pulse-chased chondrogenesis in embryonic and adult skate, Leucoraja erinacea through EdU labeling and detection with Click-iT EdU Cell Proliferation Kit from Thermo Fisher [9]. Proliferating EdU+ cells can also be detected through flow cytometry with the Click-iT Plus EdU Flow Cytometry Assay Kit from Thermo Fisher [10].

Cell Proliferation Assays and Cell Viability Assays figure 2
Figure 2. An example FACS trace of BrdU vs. 7-AAD. Courtesy and © Becton, Dickinson and Company. Reprinted with permission.

BrdU itself may affect the cell physiology. It can promote transcription factor- and chemical-induced reprogramming [11] and may also alter DNA structure [12].

SymProteinTop three suppliers
H3-3AH3.3 histone ACell Signaling Technology 9733 (137), Abcam ab14955 (35), Active Motif 39535 (18)
MCM2minichromosome maintenance complex component 2Cell Signaling Technology 3619 (7), BD Biosciences 610700 (6), Abcam ab108935 (3)
MKI67marker of proliferation Ki-67Invitrogen MA5-14520 (797), Dako M7240 (295), Abcam ab16667 (272)
PCNAproliferating cell nuclear antigenSanta Cruz Biotechnology sc-56 (206), Abcam ab29 (100), Invitrogen 13-3900 (77)
Table 1. Proliferation markers and top cited antibodies against them among the over 60,000 formal publications in Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.
Staining for markers of proliferation

In sections of fixed animal tissues and cell populations, proliferation is usually assessed by immunostaining for specific proliferative markers, some of which are described here. Ki-67 is a nuclear protein associated with cell proliferation and ribosomal RNA transcription [13] and commonly used [14]. Traditional Ki67 antibodies are limited in that they can only be used to stain frozen, and not paraffin-embedded sections. However, novel MIB-1 antibodies, directed against a different epitope of Ki67 can also be used to stain formalin and paraffin-fixed sections, increase the application of Ki67 as a proliferative marker. Ouadah Y et al stained mouse lung cryosections with Ki-67 antibodies (Thermo Fisher 41-5698-82 and 50-5698-82) to investigate injury-induce neuroendocrine cell proliferation [8]. Nam S et al used Ki-67 staining and flow cytometry to estimate fraction of cells from 3D hydrogel culture in G0 and G1 phases of the cell cycle [15]. Hu CK et al, on the other hand, stained cells at the G2/M transition of the cell cycle in killifish embryos with Hoechst and antibodies to phospho-Histone H3 (Ser10) to assess cell proliferation [16].

Proliferating cell nuclear antigen (PCNA) is another commonly used marker of cell proliferation. It expedites DNA synthesis with DNA polymerase-δ by encircling the genome facilitating replication by holding the polymerase to the DNA. It is therefore expressed in the nucleus during DNA synthesis and as such, can be used as a marker of cell proliferation. It also plays important roles in DNA repair. Other proliferating markers include MCM2 [17].

Considerations for proliferation assays

All assays either directly or indirectly measuring DNA synthesis are intrinsically sensitive to the stage of the cell cycle. Depending on the outcome of the assay, it may, therefore, be necessary to synchronize the cells, either by serum withdrawal which accumulates cells in G1 (which may also affect viability) or chemically inhibit DNA synthesis, blocking cells in S phase, with thymidine, cytosine arabinoside, hydroxyurea or aminopterin. In addition, cell proliferation can also be measured through monitoring of cell division by using, for example, Cell Proliferation Dye eFluor™ 670 from Thermo Fisher [18].

Cell Viability Assays

Cell-based assays to measure viability can be divided mainly into three categories: those that exploit the loss of membrane integrity, those that directly measure metabolic markers, and those that assess metabolic activity. Other forms of detection exist. Crystal violet staining can check the adherence of cells and thus measure the viability of adherent cells [19, 20]. For example, Lee YR et al used crystal violet staining to assess the effects of PTEN K342/K344R mutant on the proliferation of PC3 cells [21]. Vasan N et al stained fixed MCF10A cells with crystal violet to measure cell proliferation [22].

Metabolic assays

There are now many different metabolic assays available, many of which are discussed below. These assays either involve measuring levels of important metabolic proteins such as ATP or utilize the reduction of either tetrazolium salts or resazurin dyes inside cells. The ratios of NADPH/NADP, FADH/FAD, FMNH/FMN, and NADH/NAD all increase during cellular proliferation. In the presence of these metabolic intermediate cellular dehydrogenases or reductases, tetrazolium salts get reduced to a formazan product, which can be detected by the resulting colorimetric change. Similarly, resazurin (7-Hydroxy-3H-phenoxazin-3-one 10-oxide) is a non-fluorescent blue redox dye that gets reduced to resorufin, a red fluorescent compound, giving both fluorescent and colorimetric changes. Commonly used substrates, together with some of their pros and cons, are described here:

  • MTT: MTT (3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-tetrazolium bromide) is a tetrazolium salt that gets reduced by both mitochondrial and extra-mitochondrial dehydrogenases to form insoluble blue formazan crystals, meaning a solubilization step is required before the assay can be read [23]. In addition, cells become non-viable during this assay, meaning that repeat or complementary assays cannot be carried out on the same plate of cells.
  • MTS/XTT: MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphophenyl)-2H-tetrazolium) and XTT (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) substrates are similar to MTT. However, one advantage is that the reactions are carried out intracellularly in the presence of the intermediate electron acceptor phenazine methosulfate (PMS), which enhances their sensitivity. In addition, the reduced formazan product is soluble and gets released to the culture media, removing the need for the extra solubility step that is required with MTT. However, phenol red in cell culture media, fatty acids, and serum albumin have all been reported to distort data obtained from MTS, XTT, and WST assays over prolonged incubation periods [24]. Promega RealTime Glo MT Cell Viability Assay explores the reductive environment inside cells to reduce a chemical into a suitable substrate for NanoLuc luciferase. Reynders M et al, for example, measured cell cytotoxicity and proliferation with an MTS assay [25].
  • Alamar Blue: Alamar Blue is a resazurin compound that gets reduced to resorufin and dihydroresorufin in viable cells. It can enter live cells so does not require cell lysis, and is stable in culture media. This assay has the added advantage that it can be measured in both fluorimetric and colorimetric plate readers. Silva MC et al used Thermo Fisher Alamar Blue reagent to measure the cell viability of induced pluripotent cell-derived neural progenitor cells upon treatment of tau protein degrader QC-01–175 [26]. Dominy SS et al measured the viability of human neuroblastoma SH-SY5Y cells challenged with Porphyromonas gingivalis with or without gingipain inhibitors or antibiotics using Thermo Fisher Alamar Blue reagent [27].
  • WST: Water-soluble tetrazolium salts (WSTs) are cell-impermeable tetrazolium dyes that get reduced extracellularly via plasma membrane electron transport [28], and combined with the electron acceptor PMS to generate water-soluble formazan dyes. Noda S et al, for example, examined the viability of primary dental pulp stem cells with Cell Counting Kit-8 (containing WST-8 solution, also called CCK-8 assay), from Dojindo Laboratories [29]. L Zhao et al used the same kit to measure tumor cell proliferation in culture [30]. Luther A et al used a similar WST-8 kit, but from MilliporeSigma, to measure the cytotoxicity of prospective antibiotics on HeLa and HEP G2 cells [31].

There are numerously available assays that measure ATP levels as an output of overall cell health. When cells begin to undergo apoptosis or lose membrane integrity, ATP stocks become depleted through the activity of ATPases that concurrently prevent any new ATP synthesis. This leads to a rapid depletion of intracellular ATP levels. Luminescent ATP assays (such as Promega’s CellTiter-Glo) function by lysing cells to release ATP stores, while concurrently inhibiting ATPases. Luciferase catalyzes the oxidation of luciferin to oxyluciferin in the presence of magnesium and ATP, resulting in a luminescent signal that directly correlates with the intracellular ATP concentration [32, 33].

Limitations and considerations when using metabolic assays

There are a number of decisions to make when selecting the appropriate metabolic assay for your needs. Each of the substrates listed above, and related ones not covered here, have their distinct advantages and disadvantages when directly compared. Assay sensitivity, noise to signal ratio, ease of use, and reagent stability are all factors to consider. An additional important consideration with metabolic assays is that the reduction of these substrates is impacted by changes in intracellular metabolic activity that have no direct effect on overall cell viability; therefore the question you are trying to answer will play a key role in selecting the appropriate assays.

Measures of membrane integrity

All viability assays in this category rely on the breakdown of the cell membrane during the loss of viability to either allow macromolecules to enter the cell, or allow intracellular proteins to be secreted to the culture media.

  • LDH: Lactate dehydrogenase is a ubiquitous, stable cytoplasmic enzyme that converts lactate to pyruvate. If the cell membrane has been damaged, LDH, and therefore, its enzymatic activity is released from cells and can be detected in cell culture media. During the conversion of lactate to pyruvate, NAD+ gets reduced to NADH/H+. LDH-based viability assays capitalize on the formation of the free hydrogen ion by catalyzing the transfer of H+ from NADH/H+ to the tetrazolium salt INT (2-(4-iodophenyl)-3-(4-nitrophenyl)-5-phenyltetrazolium chloride), reducing it to a red formazan dye [34], or conversion of resazurin to the fluorescent form resorufin in Promega CytoTox-ONE assay [35]. Persson EK et al used Pierce LDH cytotoxicity assay kit to measure necrotic cell death due to Charcot-Leyden crystals [36].
  • Trypan Blue: Staining a cell suspension with trypan blue is one of the oldest and simplest viability assays [37, 38]. In a healthy, viable cell, the intact membrane will prevent trypan blue from entering cells. In dead or dying cells, trypan blue will enter the cell, staining it blue. This method was traditionally quantified manually using microscopes and hemocytometers, making it very labor-intensive. However, the recent availability of affordable automated cell counters, for example, Bio-Rad TC20 automated cell counter [38] or Vi-CELL™ automated cell viability analyzer and counter from Beckman Coulter [39], makes this assay less time consuming and more accurate than previously. Other similar, but fluorescent dyes can be used as well. For example, Aizarani N et al identifed viable cells with a cell sorter through Zombie Green from BioLegend [40].
  • Calcein-AM: Calcein-acetoxymethylester is a non-fluorescent dye that is used in both cell viability and apoptosis assays, for example, for detecting the cytotoxicity of Abeta peptide and its oligomers [41]. It is lipophilic, allowing easy passage through the cell membrane. Once inside the cell, intracellular esterases cleave the ester bonds of the acetomethoxy group, resulting in the formation of a fluorescent anionic and hydrophilic calcein dye, which gets trapped inside the cell. Non-viable cells do not contain active esterases, allowing this assay to be used as a measure of viability. Cu2+, Co2+, Fe3+, Mn2+, and Ni2+ quench the fluorescent signal from calcein at physiological pH, which means care must be taken to select the appropriate cell culture media.
  • Propidium Iodide/7-AAD: These intercalating agents are frequently used to study the cell cycle as discussed above. However, since they are membrane-impermeable, they are excluded from viable cells. This means that the fluorescence signal emitted by PI or 7-AAD in non-viable cells can be measured either by fluorescence microscopy [42] or FACS analysis [43, 44]. For example, Nortley R et al evaluated the pericyte cell death with 7.5 uM propidium iodide [45]. Capello M et al used IncuCyte™ Cytotox Green Reagent from Essen Bioscience to measure the cell death among cultured cancer cells [46].
  • Cell-impermeable DNA-binding dyes such as DRAQ7 from Abcam [47] or SYTOX from Thermo Fisher. These dyes enter cells through compromised cell memebranes and display strong fluorescence upon binding with DNA. For example, Samir P et al monitored cell death in cultured bone-marrow-derived macrophages in real time with SYTOX Green staining under a two-colour IncuCyte Zoom incubator imaging system from Essen Biosciences [48].
Results from a Labome Survey of Formal Publications

This section is provided by Labome to help guide researchers to identify most suited cell proliferation and cell viability assay kits. Labome surveys formal publications. Table 2 lists the major suppliers for reagents/kits used in the cell-based assays. Some of the applications are enumerated here. Zeng Q et al measured breast cancer cell growth with an MTT cell proliferation kit from Roche [49]. Nam S et al measured cell proliferation in 3D-hydrogel with EdU from Thermo Fisher Scientific [15]. Ombrato L et al assessed the in vitro proliferation of MMTV–PyMT actin–GFP cells in 2D co-culture with MACS-sorted EPCAM+ and Ly6G+ mouse cells using Click-iT Plus EdU Flow Cytometry Assay Kit from Thermo Fisher in a flow cytometer [50]. Silvestre-Roig C et al assayed the cell viability of smooth muscle cells incubated with isolated NETs with propidium iodide, Vybrant MTT cell proliferation assay from Thermo Fisher, and for live imaging, calcein AM from Thermo Fisher [51]. Genet G et al measured the proliferation of HUVEC cells in response to VEGF-A with the xCELLigence RTCA DP analyzer from ACEA Biosciences [3]. Herb M et al assessed the viability of macrophages in culture with the CyQUANT Direct Cell Proliferation Assay from Thermo Fisher Scientific [52].

supplierkitmethodssample references
AbcamDRAQ7cytometry or flow cytometry [47]
Abcamcalcein AMflow cytometry [53]
ACEA BiosciencesxCELLigence RTCA DP analyzerlabel-free [3]
BD PharmingenBrdU Flow Kitflow cytometry [54]
BioLegendcarboxyfluorescein succinimidyl esterflow cytometry [55]
BioLegendTag-it-violetflow cytometry [43]
BioLegendZombie Aquaflow cytometry, immunocytochemistry [56-58]
BioLegendZombie GreenFACS [40]
BioLegendZombie NIRFACS [59]
Dojindo LaboratoriesCell Counting Kit-8 Greencolorimetry [29]
Essen BioscienceIncuCyte Cytotox Green [46]
MilliporeSigmaBrdUimmunohistochemistry [8]
MilliporeSigmaCell Counting Kit-8 [31]
MilliporeSigmaMTT [49, 60]
PromegaCellTiter 96 AQueouscell imaging [43, 46, 61, 62]
PromegaCellTiter-Gloluminescent assay [47, 63]
PromegaCellTiter-Glo 3Dluminescent assay [64]
PromegaCytoTox 96 [65, 66]
PromegaCytoTox-ONE [35]
PromegaRealTime-Glo MT [67, 68]
RocheWST-1 [69]
Thermo FisherAlamar Blueimaging [26, 27]
Thermo Fishercalcein AMimaging [51]
Thermo FisherCell Proliferation Dye e670flow cytometry [18]
Thermo FisherCellTrace Violet Cell Proliferationflow cytometry [54]
Thermo FisherClick-iT Eduimmunochemistry, flow cytometry [8, 70]
Thermo FisherCyQUANT Direct Cell Proliferation Assaycell imaging [71, 72]
Thermo FisherLIVE/DEAD Fixable Blue Dead Cell Stain Kitflow cytometry [73]
Thermo FisherSYTOXcell imaging [48, 64]
Thermo FisherTrypan Bluecell imaging [74]
Thermo FisherVybrant MTTcolorimetry [75]
Table 3. Major cell proliferation and viability reagents/kits based on the Labome survey of formal publications.
Acknowledgement

This article is derived from an earlier version of an article authored by Dr. Laura Cobb "Cell-Based Assays: the Cell Cycle, Cell Proliferation and Cell Death", written in February 2013.

References
  1. De Cecco M, Ito T, Petrashen A, Elias A, Skvir N, Criscione S, et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature. 2019;566:73-78 pubmed publisher
  2. Wiechmann S, Maisonneuve P, Grebbin B, Hoffmeister M, Kaulich M, Clevers H, et al. Conformation-specific inhibitors of activated Ras GTPases reveal limited Ras dependency of patient-derived cancer organoids. J Biol Chem. 2020;: pubmed publisher
  3. Genet G, Boyé K, Mathivet T, Ola R, Zhang F, Dubrac A, et al. Endophilin-A2 dependent VEGFR2 endocytosis promotes sprouting angiogenesis. Nat Commun. 2019;10:2350 pubmed publisher
  4. Meharena H, Marco A, Dileep V, Lockshin E, Akatsu G, Mullahoo J, et al. Down-syndrome-induced senescence disrupts the nuclear architecture of neural progenitors. Cell Stem Cell. 2022;29:116-130.e7 pubmed publisher
  5. Maun H, Jackman J, Choy D, Loyet K, Staton T, Jia G, et al. An Allosteric Anti-tryptase Antibody for the Treatment of Mast Cell-Mediated Severe Asthma. Cell. 2019;179:417-431.e19 pubmed publisher
  6. Salic A, Mitchison T. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc Natl Acad Sci U S A. 2008;105:2415-20 pubmed publisher
  7. de Morrée A, Klein J, Gan Q, Farup J, Urtasun A, Kanugovi A, et al. Alternative polyadenylation of Pax3 controls muscle stem cell fate and muscle function. Science. 2019;366:734-738 pubmed publisher
  8. Ouadah Y, Rojas E, Riordan D, Capostagno S, Kuo C, Krasnow M. Rare Pulmonary Neuroendocrine Cells Are Stem Cells Regulated by Rb, p53, and Notch. Cell. 2019;179:403-416.e23 pubmed publisher
  9. Marconi A, Hancock Ronemus A, Gillis J. Adult chondrogenesis and spontaneous cartilage repair in the skate, Leucoraja erinacea. elife. 2020;9: pubmed publisher
  10. de Laval B, Maurizio J, Kandalla P, Brisou G, Simonnet L, Huber C, et al. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells. Cell Stem Cell. 2020;26:657-674.e8 pubmed publisher
  11. Long Y, Wang M, Gu H, Xie X. Bromodeoxyuridine promotes full-chemical induction of mouse pluripotent stem cells. Cell Res. 2015;25:1171-4 pubmed publisher
  12. Taupin P. BrdU immunohistochemistry for studying adult neurogenesis: paradigms, pitfalls, limitations, and validation. Brain Res Rev. 2007;53:198-214 pubmed
  13. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182:311-22 pubmed
  14. Wang W, Hu C, Zeng A, Alegre D, Hu D, Gotting K, et al. Changes in regeneration-responsive enhancers shape regenerative capacities in vertebrates. Science. 2020;369: pubmed publisher
  15. Nam S, Gupta V, Lee H, Lee J, Wisdom K, Varma S, et al. Cell cycle progression in confining microenvironments is regulated by a growth-responsive TRPV4-PI3K/Akt-p27Kip1 signaling axis. Sci Adv. 2019;5:eaaw6171 pubmed publisher
  16. Hu C, Wang W, Brind Amour J, Singh P, Reeves G, Lorincz M, et al. Vertebrate diapause preserves organisms long term through Polycomb complex members. Science. 2020;367:870-874 pubmed publisher
  17. Coolen M, Labusch M, Mannioui A, Bally Cuif L. Mosaic Heterochrony in Neural Progenitors Sustains Accelerated Brain Growth and Neurogenesis in the Juvenile Killifish N. furzeri. Curr Biol. 2020;30:736-745.e4 pubmed publisher
  18. Divine R, Dang H, Ueda G, Fallas J, Vulovic I, Sheffler W, et al. Designed proteins assemble antibodies into modular nanocages. Science. 2021;372: pubmed publisher
  19. Li L, Lai F, Hu X, Liu B, Lu X, Lin Z, et al. Multifaceted SOX2-chromatin interaction underpins pluripotency progression in early embryos. Science. 2023;382:eadi5516 pubmed publisher
  20. Feoktistova M, Geserick P, Leverkus M. Crystal Violet Assay for Determining Viability of Cultured Cells. Cold Spring Harb Protoc. 2016;2016:pdb.prot087379 pubmed publisher
  21. Lee Y, Chen M, Lee J, Zhang J, Lin S, Fu T, et al. Reactivation of PTEN tumor suppressor for cancer treatment through inhibition of a MYC-WWP1 inhibitory pathway. Science. 2019;364: pubmed publisher
  22. VASAN N, Razavi P, Johnson J, Shao H, Shah H, Antoine A, et al. Double PIK3CA mutations in cis increase oncogenicity and sensitivity to PI3Kα inhibitors. Science. 2019;366:714-723 pubmed publisher
  23. Liu Y, Peterson D, Kimura H, Schubert D. Mechanism of cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J Neurochem. 1997;69:581-93 pubmed
  24. Huang K, Chen Y, Walker A. Inaccuracies in MTS assays: major distorting effects of medium, serum albumin, and fatty acids. Biotechniques. 2004;37:406, 408, 410-2 pubmed
  25. Reynders M, Matsuura B, Bérouti M, Simoneschi D, Marzio A, Pagano M, et al. PHOTACs enable optical control of protein degradation. Sci Adv. 2020;6:eaay5064 pubmed publisher
  26. Silva M, Ferguson F, Cai Q, Donovan K, Nandi G, Patnaik D, et al. Targeted degradation of aberrant tau in frontotemporal dementia patient-derived neuronal cell models. elife. 2019;8: pubmed publisher
  27. Dominy S, LYNCH C, Ermini F, Benedyk M, Marczyk A, Konradi A, et al. Porphyromonas gingivalis in Alzheimer's disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 2019;5:eaau3333 pubmed publisher
  28. Berridge M, Herst P, Tan A. Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnol Annu Rev. 2005;11:127-52 pubmed
  29. Noda S, Kawashima N, Yamamoto M, Hashimoto K, Nara K, Sekiya I, et al. Effect of cell culture density on dental pulp-derived mesenchymal stem cells with reference to osteogenic differentiation. Sci Rep. 2019;9:5430 pubmed publisher
  30. Zhao L, Han X, Lu J, McEachern D, Wang S. A highly potent PROTAC androgen receptor (AR) degrader ARD-61 effectively inhibits AR-positive breast cancer cell growth in vitro and tumor growth in vivo. Neoplasia. 2020;22:522-532 pubmed publisher
  31. Luther A, Urfer M, Zahn M, Müller M, Wang S, Mondal M, et al. Chimeric peptidomimetic antibiotics against Gram-negative bacteria. Nature. 2019;: pubmed publisher
  32. Lundin A, Hasenson M, Persson J, Pousette A. Estimation of biomass in growing cell lines by adenosine triphosphate assay. Methods Enzymol. 1986;133:27-42 pubmed
  33. Crouch S, Kozlowski R, Slater K, Fletcher J. The use of ATP bioluminescence as a measure of cell proliferation and cytotoxicity. J Immunol Methods. 1993;160:81-8 pubmed
  34. Decker T, Lohmann Matthes M. A quick and simple method for the quantitation of lactate dehydrogenase release in measurements of cellular cytotoxicity and tumor necrosis factor (TNF) activity. J Immunol Methods. 1988;115:61-9 pubmed
  35. Choi S, Bylykbashi E, Chatila Z, Lee S, Pulli B, Clemenson G, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer's mouse model. Science. 2018;361: pubmed publisher
  36. Persson E, Verstraete K, Heyndrickx I, Gevaert E, Aegerter H, Percier J, et al. Protein crystallization promotes type 2 immunity and is reversible by antibody treatment. Science. 2019;364: pubmed publisher
  37. Karthaus W, Hofree M, Choi D, Linton E, Turkekul M, Bejnood A, et al. Regenerative potential of prostate luminal cells revealed by single-cell analysis. Science. 2020;368:497-505 pubmed publisher
  38. Gate D, Saligrama N, Leventhal O, Yang A, Unger M, Middeldorp J, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer's disease. Nature. 2020;577:399-404 pubmed publisher
  39. Boettcher S, Miller P, Sharma R, McConkey M, Leventhal M, Krivtsov A, et al. A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science. 2019;365:599-604 pubmed publisher
  40. Aizarani N, Saviano A, Sagar -, Mailly L, Durand S, Herman J, et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature. 2019;572:199-204 pubmed publisher
  41. Robbins J, Perfect L, Ribe E, Maresca M, Dangla Valls A, Foster E, et al. Clusterin Is Required for β-Amyloid Toxicity in Human iPSC-Derived Neurons. Front Neurosci. 2018;12:504 pubmed publisher
  42. Yasuda S, Tsuchiya H, Kaiho A, Guo Q, Ikeuchi K, Endo A, et al. Stress- and ubiquitylation-dependent phase separation of the proteasome. Nature. 2020;578:296-300 pubmed publisher
  43. Chew H, De Lima P, Gonzalez Cruz J, Banushi B, Echejoh G, Hu L, et al. Endocytosis Inhibition in Humans to Improve Responses to ADCC-Mediating Antibodies. Cell. 2020;180:895-914.e27 pubmed publisher
  44. Schneider D, Xiong Y, Wu D, Hu P, Alabanza L, Steimle B, et al. Trispecific CD19-CD20-CD22-targeting duoCAR-T cells eliminate antigen-heterogeneous B cell tumors in preclinical models. Sci Transl Med. 2021;13: pubmed publisher
  45. Nortley R, Korte N, Izquierdo P, Hirunpattarasilp C, Mishra A, Jaunmuktane Z, et al. Amyloid β oligomers constrict human capillaries in Alzheimer's disease via signaling to pericytes. Science. 2019;: pubmed publisher
  46. Capello M, Vykoukal J, Katayama H, Bantis L, Wang H, Kundnani D, et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against complement-mediated cytotoxicity. Nat Commun. 2019;10:254 pubmed publisher
  47. Eaton J, Furst L, Ruberto R, Moosmayer D, Hilpmann A, Ryan M, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;: pubmed publisher
  48. Samir P, Kesavardhana S, Patmore D, Gingras S, Malireddi R, Karki R, et al. DDX3X acts as a live-or-die checkpoint in stressed cells by regulating NLRP3 inflammasome. Nature. 2019;573:590-594 pubmed publisher
  49. Zeng Q, Michael I, Zhang P, Saghafinia S, Knott G, Jiao W, et al. Synaptic proximity enables NMDAR signalling to promote brain metastasis. Nature. 2019;573:526-531 pubmed publisher
  50. Ombrato L, Nolan E, Kurelac I, Mavousian A, Bridgeman V, Heinze I, et al. Metastatic-niche labelling reveals parenchymal cells with stem features. Nature. 2019;572:603-608 pubmed publisher
  51. Silvestre Roig C, Braster Q, Wichapong K, LEE E, Teulon J, Berrebeh N, et al. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236-240 pubmed publisher
  52. Herb M, Gluschko A, Wiegmann K, Farid A, Wolf A, Utermöhlen O, et al. Mitochondrial reactive oxygen species enable proinflammatory signaling through disulfide linkage of NEMO. Sci Signal. 2019;12: pubmed publisher
  53. Yokota T, McCourt J, Ma F, Ren S, Li S, Kim T, et al. Type V Collagen in Scar Tissue Regulates the Size of Scar after Heart Injury. Cell. 2020;: pubmed publisher
  54. Barišić D, Stadler M, Iurlaro M, Schübeler D. Mammalian ISWI and SWI/SNF selectively mediate binding of distinct transcription factors. Nature. 2019;569:136-140 pubmed publisher
  55. Hang S, Paik D, Yao L, Kim E, Jamma T, Lu J, et al. Bile acid metabolites control TH17 and Treg cell differentiation. Nature. 2019;576:143-148 pubmed publisher
  56. Virk H, Rekas M, Biddle M, Wright A, Sousa J, Weston C, et al. Validation of antibodies for the specific detection of human TRPA1. Sci Rep. 2019;9:18500 pubmed publisher
  57. Gil Cruz C, Perez Shibayama C, De Martin A, Ronchi F, Van der Borght K, Niederer R, et al. Microbiota-derived peptide mimics drive lethal inflammatory cardiomyopathy. Science. 2019;366:881-886 pubmed publisher
  58. Rosshart S, Herz J, Vassallo B, Hunter A, Wall M, Badger J, et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science. 2019;365: pubmed publisher
  59. Xu Q, Milanez Almeida P, Martins A, Radtke A, Hoehn K, Oguz C, et al. Adaptive immune responses to SARS-CoV-2 persist in the pharyngeal lymphoid tissue of children. Nat Immunol. 2023;24:186-199 pubmed publisher
  60. Liu X, Li J, Cadilha B, Markota A, Voigt C, Huang Z, et al. Epithelial-type systemic breast carcinoma cells with a restricted mesenchymal transition are a major source of metastasis. Sci Adv. 2019;5:eaav4275 pubmed publisher
  61. van der Kant R, Langness V, Herrera C, Williams D, Fong L, Leestemaker Y, et al. Cholesterol Metabolism Is a Druggable Axis that Independently Regulates Tau and Amyloid-β in iPSC-Derived Alzheimer's Disease Neurons. Cell Stem Cell. 2019;24:363-375.e9 pubmed publisher
  62. Besse A, Besse L, Kraus M, Mendez Lopez M, Bader J, Xin B, et al. Proteasome Inhibition in Multiple Myeloma: Head-to-Head Comparison of Currently Available Proteasome Inhibitors. Cell Chem Biol. 2019;26:340-351.e3 pubmed publisher
  63. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375:1254-1261 pubmed publisher
  64. Wu J, Minikes A, Gao M, Bian H, Li Y, Stockwell B, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 2019;572:402-406 pubmed publisher
  65. Ślężak T, Bailey L, Jaskolowski M, Nahotko D, Filippova E, Davydova E, et al. An engineered ultra-high affinity Fab-Protein G pair enables a modular antibody platform with multifunctional capability. Protein Sci. 2019;: pubmed publisher
  66. Cuchet Lourenço D, Eletto D, Wu C, Plagnol V, Papapietro O, CURTIS J, et al. Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation. Science. 2018;361:810-813 pubmed publisher
  67. Bellail A, Jin H, Lo H, Jung S, Hamdouchi C, Kim D, et al. Ubiquitination and degradation of SUMO1 by small-molecule degraders extends survival of mice with patient-derived tumors. Sci Transl Med. 2021;13:eabh1486 pubmed publisher
  68. Frottin F, Schueder F, Tiwary S, Gupta R, Korner R, Schlichthaerle T, et al. The nucleolus functions as a phase-separated protein quality control compartment. Science. 2019;365:342-347 pubmed publisher
  69. Lee M, Siddoway B, Kaeser G, Segota I, Rivera R, Romanow W, et al. Somatic APP gene recombination in Alzheimer's disease and normal neurons. Nature. 2018;563:639-645 pubmed publisher
  70. Shwartz Y, Gonzalez Celeiro M, Chen C, Pasolli H, Sheu S, Fan S, et al. Cell Types Promoting Goosebumps Form a Niche to Regulate Hair Follicle Stem Cells. Cell. 2020;: pubmed publisher
  71. Leone R, Zhao L, Englert J, Sun I, Oh M, Sun I, et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 2019;366:1013-1021 pubmed publisher
  72. Clark I, Gutiérrez Vázquez C, Wheeler M, Li Z, Rothhammer V, Linnerbauer M, et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science. 2021;372: pubmed publisher
  73. Rigau M, Ostrouska S, Fulford T, Johnson D, Woods K, Ruan Z, et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by γδ T cells. Science. 2020;367: pubmed publisher
  74. Siemionow M, Langa P, Brodowska S, Kozlowska K, Zalants K, Budzynska K, et al. Long-Term Protective Effect of Human Dystrophin Expressing Chimeric (DEC) Cell Therapy on Amelioration of Function of Cardiac, Respiratory and Skeletal Muscles in Duchenne Muscular Dystrophy. Stem Cell Rev Rep. 2022;: pubmed publisher
  75. Hayashi H, Mamun A, Takeyama M, Yamamura A, Zako M, Yagasaki R, et al. Activator of G-protein signaling 8 is involved in VEGF-induced choroidal neovascularization. Sci Rep. 2019;9:1560 pubmed publisher
ISSN : 2329-5139