1
|
Brozowski JM, Timoshchenko RG, Serafin DS, Allyn B, Koontz J, Rabjohns EM, Rampersad RR, Ren Y, Eudy AM, Harris TF, Abraham D, Mattox D, Rubin CT, Hilton MJ, Rubin J, Allbritton NL, Billard MJ, Tarrant TK. G protein-coupled receptor kinase 3 modulates mesenchymal stem cell proliferation and differentiation through sphingosine-1-phosphate receptor regulation. Stem Cell Res Ther 2022; 13:37. [PMID: 35093170 PMCID: PMC8800243 DOI: 10.1186/s13287-022-02715-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 12/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The bone marrow niche supports hematopoietic cell development through intimate contact with multipotent stromal mesenchymal stem cells; however, the intracellular signaling, function, and regulation of such supportive niche cells are still being defined. Our study was designed to understand how G protein receptor kinase 3 (GRK3) affects bone marrow mesenchymal stem cell function by examining primary cells from GRK3-deficient mice, which we have previously published to have a hypercellular bone marrow and leukocytosis through negative regulation of CXCL12/CXCR4 signaling. METHODS Murine GRK3-deficient bone marrow mesenchymal stromal cells were harvested and cultured to differentiate into three lineages (adipocyte, chondrocyte, and osteoblast) to confirm multipotency and compared to wild type cells. Immunoblotting, modified-TANGO experiments, and flow cytometry were used to further examine the effects of GRK3 deficiency on bone marrow mesenchymal stromal cell receptor signaling. Microcomputed tomography was used to determine trabecular and cortical bone composition of GRK3-deficient mice and standard ELISA to quantitate CXCL12 production from cellular cultures. RESULTS GRK3-deficient, bone marrow-derived mesenchymal stem cells exhibit enhanced and earlier osteogenic differentiation in vitro. The addition of a sphingosine kinase inhibitor abrogated the osteogenic proliferation and differentiation, suggesting that sphingosine-1-phosphate receptor signaling was a putative G protein-coupled receptor regulated by GRK3. Immunoblotting showed prolonged ERK1/2 signaling after stimulation with sphingosine-1-phosphate in GRK3-deficient cells, and modified-TANGO assays suggested the involvement of β-arrestin-2 in sphingosine-1-phosphate receptor internalization. CONCLUSIONS Our work suggests that GRK3 regulates sphingosine-1-phosphate receptor signaling on bone marrow mesenchymal stem cells by recruiting β-arrestin to the occupied GPCR to promote internalization, and lack of such regulation affects mesenchymal stem cell functionality.
Collapse
Affiliation(s)
- Jaime M Brozowski
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA
| | - Roman G Timoshchenko
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - D Stephen Serafin
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Brittney Allyn
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA
| | - Jessica Koontz
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Emily M Rabjohns
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA
| | - Rishi R Rampersad
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA
| | - Yinshi Ren
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC, USA
| | - Amanda M Eudy
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA
| | - Taylor F Harris
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Abraham
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Mattox
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering at Stony, Brook University, Stony Brook, NY, USA
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew J Billard
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Teresa K Tarrant
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, USA.
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, 200 Trent Dr., DUMC 3874, Durham, NC, USA.
- School of Medicine, Duke University, 152 Edwin L. Jones Building, 207 Research Drive, Durham, NC, 27710, USA.
| |
Collapse
|
2
|
Abraham DH, Anttila MM, Gallion LA, Petersen BV, Proctor A, Allbritton NL. Design of an automated capillary electrophoresis platform for single-cell analysis. Methods Enzymol 2019; 628:191-221. [PMID: 31668230 DOI: 10.1016/bs.mie.2019.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Single-cell analysis of cellular contents by highly sensitive analytical instruments is known as chemical cytometry. A chemical cytometer typically samples one cell at a time, quantifies the cellular contents of interest, and then processes and reports that data. Automation adds the potential to perform this entire sequence of events with minimal intervention, increasing throughput and repeatability. In this chapter, we discuss the design considerations for an automated capillary electrophoresis-based instrument for assay of enzymatic activity within single cells. We describe the key requirements of the microscope base and capillary electrophoresis platforms. We also provide detailed protocols and schematic designs of our cell isolation, lysis, sampling, and detection strategies. Additionally, we describe our signal processing and instrument automation workflows. The described automated system has demonstrated single-cell throughput at rates above 100cells/h and analyte limits of detection as low as 10-20mol.
Collapse
Affiliation(s)
- David H Abraham
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Matthew M Anttila
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Luke A Gallion
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Brae V Petersen
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States; Joint Department of Biomedical Engineering, University of North Carolina, Chapel and North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
3
|
Emelyanov AN, Borisova MV, Kiryanova VV. Model acupuncture point: Bone marrow-derived stromal stem cells are moved by a weak electromagnetic field. World J Stem Cells 2016; 8:342-354. [PMID: 27822341 PMCID: PMC5080641 DOI: 10.4252/wjsc.v8.i10.342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/23/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To show the existence of a structural formative role of magnetic fields (MFs) with respect to biological objects by using our proposed model of an acupoint. METHODS We introduced a magnetised 10-100 μT metal rod (needle) into culture dishes with a negatively charged working surface and observed during 24 h how cells were arranged by MFs and by electrical fields (EFs) when attached. Rat and human bone marrow-derived stromal stem cells (rBMSCs and hBMSCs), human nonadherent mononuclear blood cells, NCTCs and A172 cells, and Escherichia coli (E. coli) were evaluated. The dish containing BMSCs was defined as the model of an acupoint. rBMSCs proliferative activity affected by the needle was investigated. For investigating electromagnetic field structures, we used the gas discharge visualisation (GDV) method. RESULTS During 24 h of incubation in 50-mm culture dishes, BMSCs or the nonadherent cells accumulated into a central heap in each dish. BMSCs formed a torus (central ring) with an inner diameter of approximately 10 mm only upon the introduction of the needle in the centre of the dish. The cells did not show these effects in 35- or 90-mm culture dishes or hydrophobic dishes or rectangular cuvettes. NCTCs and A172 cells showed unstable the effects and only up to two weeks after thawing. Moreover, we observed that the appearance of these effects depended on the season. In winter, BMSCs showed no the effects. GDV experiments revealed that the resonant annular illumination gradually formed from 10 to 18-20 s in polar solutions with and without cell suspension of BMSCs, NCTCs and E. coli when using circular 50-mm dishes, stimulation at 115 V and switching of the electrode poles at 1 kHz. All these data demonstrate the resonant nature of the central ring. Significant influence of MFs on the rBMSC proliferation rate was not observed. CONCLUSION BMSCs can be moved by MFs when in the presence of a constant EF and MF, when the cells are in the responsive functional state, and when there is a resonant relationship between them.
Collapse
Affiliation(s)
- Artem N Emelyanov
- Artem N Emelyanov, Laboratory of High Laser and Magnetic Technology, North-Western State Medical University, 191015 St. Petersburg, Russia
| | - Marina V Borisova
- Artem N Emelyanov, Laboratory of High Laser and Magnetic Technology, North-Western State Medical University, 191015 St. Petersburg, Russia
| | - Vera V Kiryanova
- Artem N Emelyanov, Laboratory of High Laser and Magnetic Technology, North-Western State Medical University, 191015 St. Petersburg, Russia
| |
Collapse
|
4
|
Xu J, Huang P, Qin Y, Jiang D, Chen HY. Analysis of Intracellular Glucose at Single Cells Using Electrochemiluminescence Imaging. Anal Chem 2016; 88:4609-12. [DOI: 10.1021/acs.analchem.6b01073] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Jingjing Xu
- State Key
Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Peiyuan Huang
- State Key
Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yu Qin
- State Key
Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Dechen Jiang
- State Key
Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hong-yuan Chen
- State Key
Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
5
|
Patabadige DEW, Mickleburgh T, Ferris L, Brummer G, Culbertson AH, Culbertson CT. High‐throughput microfluidic device for single cell analysis using multiple integrated soft lithographic pumps. Electrophoresis 2016; 37:1337-44. [DOI: 10.1002/elps.201500557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 01/07/2023]
Affiliation(s)
| | - Tom Mickleburgh
- Department of Chemistry Kansas State University Manhattan KS USA
| | - Lorin Ferris
- Department of Chemistry Kansas State University Manhattan KS USA
| | - Gage Brummer
- Department of Chemistry Kansas State University Manhattan KS USA
| | | | | |
Collapse
|
6
|
Li L, Li Q, Chen P, Li Z, Chen Z, Tang B. Consecutive Gated Injection-Based Microchip Electrophoresis for Simultaneous Quantitation of Superoxide Anion and Nitric Oxide in Single PC-12 Cells. Anal Chem 2015; 88:930-6. [DOI: 10.1021/acs.analchem.5b03664] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Lu Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Qingling Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Peilin Chen
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Zhongyi Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Zhenzhen Chen
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Bo Tang
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| |
Collapse
|
7
|
|
8
|
Dickinson AJ, Meyer M, Pawlak EA, Gomez S, Jaspers I, Allbritton NL. Analysis of sphingosine kinase activity in single natural killer cells from peripheral blood. Integr Biol (Camb) 2015; 7:392-401. [PMID: 25786072 PMCID: PMC4566154 DOI: 10.1039/c5ib00007f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Sphingosine-1-phosphate (S1P), a lipid second messenger formed upon phosphorylation of sphingosine by sphingosine kinase (SK), plays a crucial role in natural killer (NK) cell proliferation, migration, and cytotoxicity. Dysregulation of the S1P pathway has been linked to a number of immune system disorders and therapeutic manipulation of the pathway has been proposed as a method of disease intervention. However, peripheral blood NK cells, as identified by surface markers (CD56(+)CD45(+)CD3(-)CD16) consist of a highly diverse population with distinct phenotypes and functions and it is unknown whether the S1P pathway is similarly diverse across peripheral blood NK cells. In this work, we measured the phosphorylation of sphingosine-fluorescein (SF) and subsequent metabolism of S1P fluorescein (S1PF) to form hexadecanoic acid fluorescein (HAF) in 111 single NK cells obtained from the peripheral blood of four healthy human subjects. The percentage of SF converted to S1PF or HAF was highly variable amongst the cells ranging from 0% to 100% (S1PF) and 0% to 97% (HAF). Subpopulations of cells with varying levels of S1PF formation and metabolism were readily identified. Across all subjects, the average percentage of SF converted to S1PF or HAF was 37 ± 36% and 12 ± 19%, respectively. NK cell metabolism of SF by the different subjects was also distinct with hierarchical clustering suggesting two possible phenotypes: low (<20%) or high (>50%) producers of S1PF. The heterogeneity of SK and downstream enzyme activity in NK cells may enable NK cells to respond effectively to a diverse array of pathogens as well as incipient tumor cells. NK cells from two subjects were also loaded with S1PF to assess the activity of S1P phosphatase (S1PP), which converts S1P to sphingosine. No NK cells (n = 41) formed sphingosine, suggesting that S1PP was minimally active in peripheral blood NK cells. In contrast to the SK activity, S1PP activity was homogeneous across the peripheral blood NK cells, suggesting a bias in the SK pathway towards proliferation and migration, activities supported by S1P.
Collapse
Affiliation(s)
| | - Megan Meyer
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Erica A. Pawlak
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Shawn Gomez
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27695, USA
| | - Ilona Jaspers
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
9
|
Shehaj L, Lazo de la Vega L, Kovarik ML. Microfluidic Chemical Cytometry for Enzyme Assays of Single Cells. Methods Mol Biol 2015; 1346:221-38. [PMID: 26542725 DOI: 10.1007/978-1-4939-2987-0_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cellular heterogeneity occurs, and should be probed, at multiple levels of cellular structure and physiology from the genome to enzyme activity. In particular, single-cell measures of protein levels are complemented by single-cell measurements of the activity of these proteins. Microfluidic assays of enzyme activity at the single-cell level combine moderate to high throughput with low dead volumes and the potential for automation. Herein, we describe the steps required to fabricate and operate a microfluidic device for chemical cytometry of fluorescent or fluorogenic reporters of enzyme activity in individual cells.
Collapse
Affiliation(s)
- Livia Shehaj
- Department of Chemistry, Trinity College, 300 Summit Street, Hartford, CT, 06106, USA
| | | | - Michelle L Kovarik
- Department of Chemistry, Trinity College, 300 Summit Street, Hartford, CT, 06106, USA.
| |
Collapse
|
10
|
Metto EC, Evans K, Barney P, Culbertson AH, Gunasekara DB, Caruso G, Hulvey MK, da Silva JAF, Lunte SM, Culbertson CT. An integrated microfluidic device for monitoring changes in nitric oxide production in single T-lymphocyte (Jurkat) cells. Anal Chem 2013; 85:10188-95. [PMID: 24010877 PMCID: PMC3951964 DOI: 10.1021/ac401665u] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A considerable amount of attention has been focused on the analysis of single cells in an effort to better understand cell heterogeneity in cancer and neurodegenerative diseases. Although microfluidic devices have several advantages for single cell analysis, few papers have actually demonstrated the ability of these devices to monitor chemical changes in perturbed biological systems. In this paper, a new microfluidic channel manifold is described that integrates cell transport, lysis, injection, electrophoretic separation, and fluorescence detection into a single device, making it possible to analyze individual cells at a rate of 10 cells/min in an automated fashion. The system was employed to measure nitric oxide (NO) production in single T-lymphocytes (Jurkat cells) using a fluorescent marker, 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM DA). The cells were also labeled with 6-carboxyfluorescein diacetate (6-CFDA) as an internal standard. The NO production by control cells was compared to that of cells stimulated using lipopolysaccharide (LPS), which is known to cause the expression of inducible nitric oxide synthase (iNOS) in immune-type cells. Statistical analysis of the resulting electropherograms from a population of cells indicated a 2-fold increase in NO production in the induced cells. These results compare nicely to a recently published bulk cell analysis of NO.
Collapse
Affiliation(s)
- Eve C. Metto
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, USA
| | - Karsten Evans
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, USA
| | - Patrick Barney
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, USA
| | - Anne H. Culbertson
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, USA
| | - Dulan B. Gunasekara
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, USA
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66047, USA
| | - Giuseppe Caruso
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66047, USA
- Department of Chemical Science, Section of Biochemistry and Molecular Biology, The University of Catania, Italy
| | - Matthew K. Hulvey
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66047, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, USA
- Akermin, Inc. St. Louis, Missouri 63132, USA
| | - Jose Alberto Fracassi da Silva
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66047, USA
- Institute of Chemistry, State University of Campinas, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Bioanalítica, INCTBio
| | - Susan M. Lunte
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, USA
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66047, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, USA
| | | |
Collapse
|
11
|
A comparative analysis of the ubiquitination kinetics of multiple degrons to identify an ideal targeting sequence for a proteasome reporter. PLoS One 2013; 8:e78082. [PMID: 24205101 PMCID: PMC3812159 DOI: 10.1371/journal.pone.0078082] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 09/09/2013] [Indexed: 01/03/2023] Open
Abstract
The ubiquitin proteasome system (UPS) is the primary pathway responsible for the recognition and degradation of misfolded, damaged, or tightly regulated proteins. The conjugation of a polyubiquitin chain, or polyubiquitination, to a target protein requires an increasingly diverse cascade of enzymes culminating with the E3 ubiquitin ligases. Protein recognition by an E3 ligase occurs through a specific sequence of amino acids, termed a degradation sequence or degron. Recently, degrons have been incorporated into novel reporters to monitor proteasome activity; however only a limited few degrons have successfully been incorporated into such reporters. The goal of this work was to evaluate the ubiquitination kinetics of a small library of portable degrons that could eventually be incorporated into novel single cell reporters to assess proteasome activity. After an intensive literary search, eight degrons were identified from proteins recognized by a variety of E3 ubiquitin ligases and incorporated into a four component degron-based substrate to comparatively calculate ubiquitination kinetics. The mechanism of placement of multiple ubiquitins on the different degron-based substrates was assessed by comparing the data to computational models incorporating first order reaction kinetics using either multi-monoubiquitination or polyubiquitination of the degron-based substrates. A subset of three degrons was further characterized to determine the importance of the location and proximity of the ubiquitination site lysine with respect to the degron. Ultimately, this work identified three candidate portable degrons that exhibit a higher rate of ubiquitination compared to peptidase-dependent degradation, a desired trait for a proteasomal targeting motif.
Collapse
|
12
|
Kovarik ML, Shah PK, Armistead PM, Allbritton NL. Microfluidic chemical cytometry of peptide degradation in single drug-treated acute myeloid leukemia cells. Anal Chem 2013; 85:4991-7. [PMID: 23590517 PMCID: PMC3671928 DOI: 10.1021/ac4002029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microfluidic systems show great promise for single-cell analysis; however, as these technologies mature, their utility must be validated by studies of biologically relevant processes. An important biomedical application of these systems is characterization of tumor cell heterogeneity. In this work, we used a robust microfluidic platform to explore the heterogeneity of enzyme activity in single cells treated with a chemotherapeutic drug. Using chemical cytometry, we measured peptide degradation in the U937 acute myeloid leukemia (AML) cell line in the presence and absence of the aminopeptidase inhibitor Tosedostat (CHR-2797). The analysis of 99 untreated cells revealed rapid and consistent degradation of the peptide reporter within 20 min of loading. Results from drug-treated cells showed inhibited, but ongoing degradation of the reporter. Because the device operates at an average sustained throughput of 37 ± 7 cells/h, we were able to sample cells over the course of this time-dependent degradation. In data from 498 individual drug-treated cells, we found a linear dependence of degradation rate on amount of substrate loaded superimposed upon substantial heterogeneity in peptide processing in response to inhibitor treatment. Importantly, these data demonstrated the potential of microfluidic systems to sample biologically relevant analytes and time-dependent processes in large numbers of single cells.
Collapse
Affiliation(s)
- Michelle L. Kovarik
- Department of Chemistry, CB 3290, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Pavak K. Shah
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599 and North Carolina State University, Raleigh, NC 27695
| | - Paul M. Armistead
- Lineberger Comprehensive Cancer Center, CB 7295, University of North Carolina, Chapel Hill, NC 27599
| | - Nancy L. Allbritton
- Department of Chemistry, CB 3290, University of North Carolina, Chapel Hill, North Carolina 27599
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599 and North Carolina State University, Raleigh, NC 27695
- Lineberger Comprehensive Cancer Center, CB 7295, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
13
|
Dickinson AJ, Armistead PM, Allbritton NL. Automated capillary electrophoresis system for fast single-cell analysis. Anal Chem 2013; 85:4797-804. [PMID: 23527995 DOI: 10.1021/ac4005887] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Capillary electrophoresis (CE) is a promising technique for single-cell analysis, but its use in biological studies has been limited by low throughput. This paper presents an automated platform employing microfabricated cell traps and a three-channel system for rapid buffer exchange for fast single-cell CE. Cells loaded with fluorescein and Oregon green were analyzed at a throughput of 3.5 cells/min with a resolution of 2.3 ± 0.6 for the fluorescein and Oregon green. Cellular protein kinase B (PKB) activity, as measured by immunofluorescence staining of phospho-PKB, was not altered, suggesting that this stress-activated kinase was not upregulated during the CE experiments and that basal cell physiology was not perturbed prior to cell lysis. The activity of sphingosine kinase (SK), which is often upregulated in cancer, was measured in leukemic cells by loading a sphingosine-fluorescein substrate into cells. Sphingosine fluorescein (SF), sphingosine-1-phosphate fluorescein (S1PF), and a third fluorescent species were identified in single cells. A single-cell throughput of 2.1 cells/min was achieved for 219 total cells. Eighty-eight percent of cells possessed upregulated SK activity, although subpopulations of cells with markedly different SK activity relative to that of the population average were readily identified. This system was capable of stable and reproducible separations of biological compounds in hundreds of adherent and nonadherent cells, enabling measurements of previously uncharacterized biological phenomena.
Collapse
Affiliation(s)
- Alexandra J Dickinson
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | | | | |
Collapse
|
14
|
Fan Y, Xu F, Huang G, Lu TJ, Xing W. Single neuron capture and axonal development in three-dimensional microscale hydrogels. LAB ON A CHIP 2012; 12:4724-4731. [PMID: 22858829 DOI: 10.1039/c2lc40312a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Autapse is an unusual type of synapse generated by a neuron on itself. The ability to monitor axonal growth of single neurons and autapse formation in three-dimensions (3D) may provide fundamental information relating to many cellular processes, such as axonal development, synaptic plasticity and neural signal transmission. However, monitoring such growth is technically challenging due to the requirement for precise capture and long-term analysis of single neurons in 3D. Herein, we present a simple two-step photolithography method to efficiently capture single cells in microscale gelatin methacrylate hydrogel rings. We applied this method to capture and culture single neurons. The results demonstrated that neural axons grew and consequently formed axonal circles, indicating that our method could be an enabling tool to analyze axonal development and autapse formation. This method holds great potential for impact in multiple areas, such as neuroscience, cancer biology, and stem cell biology.
Collapse
Affiliation(s)
- Yantao Fan
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing, China 100086
| | | | | | | | | |
Collapse
|
15
|
Cecala C, Rubakhin SS, Mitchell JW, Gillette MU, Sweedler JV. A hyphenated optical trap capillary electrophoresis laser induced native fluorescence system for single-cell chemical analysis. Analyst 2012; 137:2965-72. [PMID: 22543409 PMCID: PMC3558031 DOI: 10.1039/c2an35198f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Single-cell measurements allow a unique glimpse into cell-to-cell heterogeneity; even small changes in selected cells can have a profound impact on an organism's physiology. Here an integrated approach to single-cell chemical sampling and assay are described. Capillary electrophoresis (CE) with laser-induced native fluorescence (LINF) has the sensitivity to characterize natively fluorescent indoles and catechols within individual cells. While the separation and detection approaches are well established, the sampling and injection of individually selected cells requires new approaches. We describe an optimized system that interfaces a single-beam optical trap with CE and multichannel LINF detection. A cell is localized within the trap and then the capillary inlet is positioned near the cell using a computer-controlled micromanipulator. Hydrodynamic injection allows cell lysis to occur within the capillary inlet, followed by the CE separation and LINF detection. The use of multiple emission wavelengths allows improved analyte identification based on differences in analyte fluorescence emission profiles and migration time. The system enables injections of individual rat pinealocytes and quantification of their endogenous indoles, including serotonin, N-acetyl-serotonin, 5-hydroxyindole-3-acetic acid, tryptophol and others. The amounts detected in individual cells incubated in 5-hydroxytryptophan ranged from 10(-14) mol to 10(-16) mol, an order of magnitude higher than observed in untreated pinealocytes.
Collapse
Affiliation(s)
- Christine Cecala
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Stanislav S. Rubakhin
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Jennifer W. Mitchell
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Martha U. Gillette
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Jonathan V. Sweedler
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
16
|
Abstract
Cells are extraordinarily complex, containing thousands of different analytes with concentrations spanning at least nine orders of magnitude. Analyzing single cells instead of tissue homogenates provides unique insights into cell-to-cell heterogeneity and aids in distinguishing normal cells from pathological ones. The high sensitivity and low sample consumption of capillary and on-chip electrophoresis, when integrated with fluorescence, electrochemical, and mass spectrometric detection methods, offer an ideal toolset for examining single cells and even subcellular organelles; however, the isolation and loading of such small samples into these devices is challenging. Recent advances have addressed this issue by interfacing a variety of enhanced mechanical, microfluidic, and optical sampling techniques to capillary and on-chip electrophoresis instruments for single-cell analyses.
Collapse
Affiliation(s)
- Christine Cecala
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | |
Collapse
|
17
|
Kašička V. Recent developments in CE and CEC of peptides (2009-2011). Electrophoresis 2011; 33:48-73. [DOI: 10.1002/elps.201100419] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 09/19/2011] [Accepted: 09/20/2011] [Indexed: 12/12/2022]
|
18
|
Affiliation(s)
- Yuqing Lin
- Department of Chemistry, University of Gothenburg, S-41296, Gothenburg, Sweden
| | | | | | | |
Collapse
|
19
|
Kovarik ML, Allbritton NL. Measuring enzyme activity in single cells. Trends Biotechnol 2011; 29:222-30. [PMID: 21316781 PMCID: PMC3080453 DOI: 10.1016/j.tibtech.2011.01.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 01/04/2011] [Accepted: 01/10/2011] [Indexed: 01/09/2023]
Abstract
Seemingly identical cells can differ in their biochemical state, function and fate, and this variability plays an increasingly recognized role in organism-level outcomes. Cellular heterogeneity arises in part from variation in enzyme activity, which results from interplay between biological noise and multiple cellular processes. As a result, single-cell assays of enzyme activity, particularly those that measure product formation directly, are crucial. Recent innovations have yielded a range of techniques to obtain these data, including image-, flow- and separation-based assays. Research to date has focused on easy-to-measure glycosylases and clinically-relevant kinases. Expansion of these techniques to a wider range and larger number of enzymes will answer contemporary questions in proteomics and glycomics, specifically with respect to biological noise and cellular heterogeneity.
Collapse
Affiliation(s)
- Michelle L Kovarik
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
20
|
Ryan D, Ren K, Wu H. Single-cell assays. BIOMICROFLUIDICS 2011; 5:21501. [PMID: 21559238 PMCID: PMC3089644 DOI: 10.1063/1.3574448] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 03/11/2011] [Indexed: 05/03/2023]
Abstract
This review presents an overview of literature that describes the applications of microfluidics to assay individual cells. We quantify the content of an individual mammalian cell, so that we can understand what criteria a single-cell assay must satisfy to be successful. We put in context the justification for single-cell assays and identify the characteristics that are relevant to single-cell assays. We review the literature from the past 24 months that describe the methods that use microfabrication-conventional or otherwise-and microfluidics in particular to study individual cells, and we present our views on how an increasing emphasis on three-dimensional cell culture and the demonstration of the first chemically defined cell might impact single-cell assays.
Collapse
Affiliation(s)
- Declan Ryan
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, People's Republic of China
| | | | | |
Collapse
|
21
|
Phillips KS, Lai HH, Johnson E, Sims CE, Allbritton NL. Continuous analysis of dye-loaded, single cells on a microfluidic chip. LAB ON A CHIP 2011; 11:1333-41. [PMID: 21327264 PMCID: PMC3462073 DOI: 10.1039/c0lc00370k] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Continuous analysis of two dyes loaded into single mammalian cells using laser-based lysis combined with electrophoretic separation was developed and characterized on microfluidic chips. The devices employed hydrodynamic flow to transport cells to a junction where they were mechanically lysed by a laser-generated cavitation bubble. An electric field then attracted the analyte into a separation channel while the membranous remnants passed through the intersection towards a waste reservoir. Phosphatidylcholine (PC)-supported bilayer membrane coatings (SBMs) provided a weakly negatively charged surface and prevented cell fouling from interfering with device performance. Cell lysis using a picosecond-pulsed laser on-chip did not interfere with concurrent electrophoretic separations. The effect of device parameters on performance was evaluated. A ratio of 2 : 1 was found to be optimal for the focusing-channel : flow-channel width and 3 : 1 for the flow-channel : separation-channel width. Migration times decreased with increased electric field strengths up to 333 V cm(-1), at which point the field strength was sufficient to move unlysed cells and cellular debris into the electrophoretic channel. The migration time and full width half-maximum (FWHM) of the peaks were independent of cell velocity for velocities between 0.03 and 0.3 mm s(-1). Separation performance was independent of the exact lysis location when lysis was performed near the outlet of the focusing channel. The migration time for cell-derived fluorescein and fluorescein carboxylate was reproducible with <10% RSD. Automated cell detection and lysis were required to reduce peak FWHM variability to 30% RSD. A maximum throughput of 30 cells min(-1) was achieved. Device stability was demonstrated by analyzing 600 single cells over a 2 h time span.
Collapse
Affiliation(s)
- K. Scott Phillips
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Hsuan Hong Lai
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Emily Johnson
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, 27599, USA and North Carolina State University, Raleigh, NC, 27695, USA. Fax: +1 919 843 7825
| |
Collapse
|
22
|
Abstract
The intracellular levels and spatial localizations of metabolites and peptides reflect the state of a cell and its relationship to its surrounding environment. Moreover, the amounts and dynamics of metabolites and peptides are indicative of normal or pathological cellular conditions. Here we highlight established and evolving strategies for characterizing the metabolome and peptidome of single cells. Focused studies of the chemical composition of individual cells and functionally defined groups of cells promise to provide a greater understanding of cell fate, function and homeostatic balance. Single-cell bioanalytical microanalysis has also become increasingly valuable for examining cellular heterogeneity, particularly in the fields of neuroscience, stem cell biology and developmental biology.
Collapse
Affiliation(s)
- Stanislav S. Rubakhin
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign
| | - Elena V. Romanova
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign
| | - Peter Nemes
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign
| | - Jonathan V. Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign
| |
Collapse
|