1
|
Dominguez SR, Doan PN, Rivera-Chávez F. The intersection between host-pathogen interactions and metabolism during Vibrio cholerae infection. Curr Opin Microbiol 2024; 77:102421. [PMID: 38215547 DOI: 10.1016/j.mib.2023.102421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024]
Abstract
Vibrio cholerae (V. cholerae), the etiological agent of cholera, uses cholera toxin (CT) to cause severe diarrheal disease. Cholera is still a significant cause of mortality worldwide with about half of all cholera cases and deaths occurring in children under five. Owing to the lack of cost-effective vaccination and poor vaccine efficacy in children, there is a need for alternative preventative and therapeutic strategies. Recent advances in our knowledge of the interplay between CT-induced disease and host-pathogen metabolism have opened the door for investigating how modulation of intestinal metabolism by V. cholerae during disease impacts host intestinal immunity, the gut microbiota, and pathogen-phage interactions. In this review article, we examine recent progress in our understanding of host-pathogen interactions during V. cholerae infection and discuss future work deciphering how modulation of gut metabolism during cholera intersects these processes to enable successful fecal-oral transmission of the pathogen.
Collapse
Affiliation(s)
- Sedelia R Dominguez
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Phillip N Doan
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fabian Rivera-Chávez
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Miller DR, Schaffer DK, Neely MD, McClain ES, Travis AR, Block FE, Mckenzie J, Werner EM, Armstrong L, Markov DA, Bowman AB, Ess KC, Cliffel DE, Wikswo JP. A bistable, multiport valve enables microformulators creating microclinical analyzers that reveal aberrant glutamate metabolism in astrocytes derived from a tuberous sclerosis patient. SENSORS AND ACTUATORS. B, CHEMICAL 2021; 341:129972. [PMID: 34092923 PMCID: PMC8174775 DOI: 10.1016/j.snb.2021.129972] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
There is a need for valves and pumps that operate at the microscale with precision and accuracy, are versatile in their application, and are easily fabricated. To that end, we developed a new rotary planar multiport valve to faithfully select solutions (contamination = 5.22 ± 0.06 ppb) and a rotary planar peristaltic pump to precisely control fluid delivery (flow rate = 2.4 ± 1.7 to 890 ± 77 μL/min). Both the valve and pump were implemented in a planar format amenable to single-layer soft lithographic fabrication. These planar microfluidics were actuated by a rotary motor controlled remotely by custom software. Together, these two devices constitute an innovative microformulator that was used to prepare precise, high-fidelity mixtures of up to five solutions (deviation from prescribed mixture = ±|0.02 ± 0.02| %). This system weighed less than a kilogram, occupied around 500 cm3, and generated pressures of 255 ± 47 kPa. This microformulator was then combined with an electrochemical sensor creating a microclinical analyzer (μCA) for detecting glutamate in real time. Using the chamber of the μCA as an in-line bioreactor, we compared glutamate homeostasis in human astrocytes differentiated from human-induced pluripotent stem cells (hiPSCs) from a control subject (CC-3) and a Tuberous Sclerosis Complex (TSC) patient carrying a pathogenic TSC2 mutation. When challenged with glutamate, TSC astrocytes took up less glutamate than control cells. These data validate the analytical power of the μCA and the utility of the microformulator by leveraging it to assess disease-related alterations in cellular homeostasis.
Collapse
Affiliation(s)
- Dusty R. Miller
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - David K. Schaffer
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, U.S.A
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - M. Diana Neely
- Department of Pediatrics, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, U.S.A
| | - Ethan S. McClain
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Adam R. Travis
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Frank E. Block
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Jennifer Mckenzie
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Erik M. Werner
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Laura Armstrong
- Department of Pediatrics, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, U.S.A
| | - Dmitry A. Markov
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, U.S.A
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - Aaron B. Bowman
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, U.S.A
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, U.S.A
| | - Kevin C. Ess
- Department of Pediatrics, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, U.S.A
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, U.S.A
| | - David E. Cliffel
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, U.S.A
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, U.S.A
| | - John P. Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, U.S.A
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, U.S.A
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, U.S.A
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37240, U.S.A
| |
Collapse
|
3
|
Cho JY, Liu R, Macbeth JC, Hsiao A. The Interface of Vibrio cholerae and the Gut Microbiome. Gut Microbes 2021; 13:1937015. [PMID: 34180341 PMCID: PMC8244777 DOI: 10.1080/19490976.2021.1937015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 02/04/2023] Open
Abstract
The bacterium Vibrio cholerae is the etiologic agent of the severe human diarrheal disease cholera. The gut microbiome, or the native community of microorganisms found in the human gastrointestinal tract, is increasingly being recognized as a factor in driving susceptibility to infection, in vivo fitness, and host interactions of this pathogen. Here, we review a subset of the emerging studies in how gut microbiome structure and microbial function are able to drive V. cholerae virulence gene regulation, metabolism, and modulate host immune responses to cholera infection and vaccination. Improved mechanistic understanding of commensal-pathogen interactions offers new perspectives in the design of prophylactic and therapeutic approaches for cholera control.
Collapse
Affiliation(s)
- Jennifer Y. Cho
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Department of Biochemistry, University of California, Riverside, California, USA
| | - Rui Liu
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, California, USA
| | - John C. Macbeth
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Ansel Hsiao
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| |
Collapse
|
4
|
Cross-Kingdom Activation of Vibrio Toxins by ADP-Ribosylation Factor Family GTPases. J Bacteriol 2020; 202:JB.00278-20. [PMID: 32900828 DOI: 10.1128/jb.00278-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic Vibrio species use many different approaches to subvert, attack, and undermine the host response. The toxins they produce are often responsible for the devastating effects associated with their diseases. These toxins target a variety of host proteins, which leads to deleterious effects, including dissolution of cell organelle integrity and inhibition of protein secretion. Becoming increasingly prevalent as cofactors for Vibrio toxins are proteins of the small GTPase families. ADP-ribosylation factor small GTPases (ARFs) in particular are emerging as a common host cofactor necessary for full activation of Vibrio toxins. While ARFs are not the direct target of Vibrio cholerae cholera toxin (CT), ARF binding is required for its optimal activity as an ADP-ribosyltransferase. The makes caterpillars floppy (MCF)-like and the domain X (DmX) effectors of the Vibrio vulnificus multifunctional autoprocessing repeats-in-toxin (MARTX) toxin also both require ARFs to initiate autoprocessing and activation as independent effectors. ARFs are ubiquitously expressed in eukaryotes and are key regulators of many cellular processes, and as such they are ideal cofactors for Vibrio pathogens that infect many host species. In this review, we cover in detail the known Vibrio toxins that use ARFs as cross-kingdom activators to both stimulate and optimize their activity. We further discuss how these contrast to toxins and effectors from other bacterial species that coactivate, stimulate, or directly modify host ARFs as their mechanisms of action.
Collapse
|
5
|
Cholera toxin promotes pathogen acquisition of host-derived nutrients. Nature 2019; 572:244-248. [PMID: 31367037 PMCID: PMC6727848 DOI: 10.1038/s41586-019-1453-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 06/24/2019] [Indexed: 11/08/2022]
Abstract
Vibrio cholerae is the causative agent of cholera, a
potentially lethal enteric bacterial infection1. Cholera toxin (CT) is required for
V. cholerae to cause severe disease and is also thought to
promote transmission of the organism in that victims can shed many liters of
diarrheal fluid that typically contains in excess of 1011 organisms
per liter. How the pathogen is able to reach such high concentrations in the
intestine during infection remains poorly understood. Here we show that
CT-mediated disease enhances pathogen growth and induces a distinct V.
cholerae transcriptome signature that is indicative an
iron-depleted gut niche. During infection, bacterial pathogens need to acquire
iron, a nutrient essential for growth2. The majority of iron in the mammalian host resides in a
chelated form within the porphyrin structure of heme, and V.
cholerae genetically encodes the ability to utilize heme as a
source of iron3. We show that
V. cholerae heme and vibriobactin
utilization genes confer a growth advantage to the pathogen only when CT is
produced. Furthermore, CT-induced capillary congestion pathology in the terminal
ileum correlated with an increased bioavailability of luminal heme. CT-induced
disease in the ileum also led to increased luminal concentrations of long-chain
fatty acids (LCFAs) and L-lactate metabolites, as well as upregulation of
V. cholerae iron-sulfur cluster-containing TCA cycle enzyme
genes. Genetic analysis of V. cholerae suggested that heme and
LCFA uptake-dependent growth of V. cholerae occurs during
infection but only in a strain capable of producing CT in vivo.
We conclude that CT-induced disease creates an iron-depleted metabolic niche in
the gut that selectively promotes the explosive growth of this pathogen through
acquisition of host-derived heme and fatty acids as nutrients.
Collapse
|
6
|
Davis AN, Travis AR, Miller DR, Cliffel DE. Multianalyte Physiological Microanalytical Devices. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:93-111. [PMID: 28605606 PMCID: PMC9235322 DOI: 10.1146/annurev-anchem-061516-045334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Advances in scientific instrumentation have allowed experimentalists to evaluate well-known systems in new ways and to gain insight into previously unexplored or poorly understood phenomena. Within the growing field of multianalyte physiometry (MAP), microphysiometers are being developed that are capable of electrochemically measuring changes in the concentration of various metabolites in real time. By simultaneously quantifying multiple analytes, these devices have begun to unravel the complex pathways that govern biological responses to ischemia and oxidative stress while contributing to basic scientific discoveries in bioenergetics and neurology. Patients and clinicians have also benefited from the highly translational nature of MAP, and the continued expansion of the repertoire of analytes that can be measured with multianalyte microphysiometers will undoubtedly play a role in the automation and personalization of medicine. This is perhaps most evident with the recent advent of fully integrated noninvasive sensor arrays that can continuously monitor changes in analytes linked to specific disease states and deliver a therapeutic agent as required without the need for patient action.
Collapse
Affiliation(s)
- Anna Nix Davis
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235;
| | - Adam R Travis
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235;
| | - Dusty R Miller
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235;
| | - David E Cliffel
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235;
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee 37235
| |
Collapse
|
7
|
Hutson MS, Alexander PG, Allwardt V, Aronoff DM, Bruner-Tran KL, Cliffel DE, Davidson JM, Gough A, Markov DA, McCawley LJ, McKenzie JR, McLean JA, Osteen KG, Pensabene V, Samson PC, Senutovitch NK, Sherrod SD, Shotwell MS, Taylor DL, Tetz LM, Tuan RS, Vernetti LA, Wikswo JP. Organs-on-Chips as Bridges for Predictive Toxicology. ACTA ACUST UNITED AC 2016. [DOI: 10.1089/aivt.2016.0003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- M. Shane Hutson
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Physics & Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Peter G. Alexander
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Vanessa Allwardt
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
| | - David M. Aronoff
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Infectious Diseases, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kaylon L. Bruner-Tran
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David E. Cliffel
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Jeffrey M. Davidson
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Research Service, Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Albert Gough
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dmitry A. Markov
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa J. McCawley
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jennifer R. McKenzie
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - John A. McLean
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Kevin G. Osteen
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
- Research Service, Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Virginia Pensabene
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Philip C. Samson
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Physics & Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Nina K. Senutovitch
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stacy D. Sherrod
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Matthew S. Shotwell
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - D. Lansing Taylor
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lauren M. Tetz
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Infectious Diseases, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rocky S. Tuan
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Bioengineering, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Center for Cellular and Molecular Engineering, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Center for Military Medicine Research, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Lawrence A. Vernetti
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John P. Wikswo
- Vanderbilt-Pittsburgh Resource for Organotypic Models for Predictive Toxicology, Vanderbilt University, Nashville, Tennessee, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Vanderbilt Institute for Integrative Biosystems Research & Education, Vanderbilt University, Nashville, Tennessee
- Department of Physics & Astronomy, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Department of Molecular Physiology & Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
8
|
Kimmel DW, Rogers LM, Aronoff DM, Cliffel DE. Prostaglandin E2 Regulation of Macrophage Innate Immunity. Chem Res Toxicol 2015; 29:19-25. [PMID: 26656203 DOI: 10.1021/acs.chemrestox.5b00322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Globally, maternal and fetal health is greatly impacted by extraplacental inflammation. Group B Streptococcus (GBS), a leading cause of chorioamnionitis, is thought to take advantage of the uterine environment during pregnancy in order to cause inflammation and infection. In this study, we demonstrate the metabolic changes of murine macrophages caused by GBS exposure. GBS alone prompted a delayed increase in lactate production, highlighting its ability to redirect macrophage metabolism from aerobic to anaerobic respiration. This production of lactate is thought to aid in the development and propagation of GBS throughout the surrounding tissue. Additionally, this study shows that PGE2 priming was able to exacerbate lactate production, shown by the rapid and substantial lactate increases seen upon GBS exposure. These data provide a novel model to study the role of GBS exposure to macrophages with and without PGE2 priming.
Collapse
Affiliation(s)
| | - Lisa M Rogers
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - David M Aronoff
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University , Nashville, Tennessee 37232, United States
| | | |
Collapse
|
9
|
McKenzie JR, Cognata AC, Davis AN, Wikswo JP, Cliffel DE. Real-Time Monitoring of Cellular Bioenergetics with a Multianalyte Screen-Printed Electrode. Anal Chem 2015; 87:7857-64. [PMID: 26125545 DOI: 10.1021/acs.analchem.5b01533] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Real-time monitoring of changes to cellular bioenergetics can provide new insights into mechanisms of action for disease and toxicity. This work describes the development of a multianalyte screen-printed electrode for the detection of analytes central to cellular bioenergetics: glucose, lactate, oxygen, and pH. Platinum screen-printed electrodes were designed in-house and printed by Pine Research Instrumentation. Electrochemical plating techniques were used to form quasi-reference and pH electrodes. A Dimatix materials inkjet printer was used to deposit enzyme and polymer films to form sensors for glucose, lactate, and oxygen. These sensors were evaluated in bulk solution and microfluidic environments, and they were found to behave reproducibly and possess a lifetime of up to 6 weeks. Linear ranges and limits of detection for enzyme-based sensors were found to have an inverse relationship with enzyme loading, and iridium oxide pH sensors were found to have super-Nernstian responses. Preliminary measurements where the sensor was enclosed within a microfluidic channel with RAW 264.7 macrophages were performed to demonstrate the sensors' capabilities for performing real-time microphysiometry measurements.
Collapse
Affiliation(s)
- Jennifer R McKenzie
- †Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,‡Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Andrew C Cognata
- †Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Anna N Davis
- †Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - John P Wikswo
- ‡Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee 37235, United States.,§Departments of Physics and Astronomy, Biomedical Engineering, and Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - David E Cliffel
- †Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,‡Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
10
|
Lima EA, Snider RM, Reiserer RS, McKenzie JR, Kimmel DW, Eklund SE, Wikswo JP, Cliffel DE. Multichamber Multipotentiostat System for Cellular Microphysiometry. SENSORS AND ACTUATORS. B, CHEMICAL 2014; 204:536-543. [PMID: 25242863 PMCID: PMC4167374 DOI: 10.1016/j.snb.2014.07.126] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Multianalyte microphysiometry is a powerful technique for studying cellular metabolic flux in real time. Monitoring several analytes concurrently in a number of individual chambers, however, requires specific instrumentation that is not available commercially in a single, compact, benchtop form at an affordable cost. We developed a multipotentiostat system capable of performing simultaneous amperometric and potentiometric measurements in up to eight individual chambers. The modular design and custom LabVIEW™ control software provide flexibility and allow for expansion and modification to suit different experimental conditions. Superior accuracy is achieved when operating the instrument in a standalone configuration; however, measurements performed in conjunction with a previously developed multianalyte microphysiometer have shown low levels of crosstalk as well. Calibrations and experiments with primary and immortalized cell cultures demonstrate the performance of the instrument and its capabilities.
Collapse
Affiliation(s)
- Eduardo A Lima
- Department of Earth, Atmospheric, and Planetary Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts. 02139, USA ; Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - Rachel M Snider
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - Ronald S Reiserer
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - Jennifer R McKenzie
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Chemistry, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - Danielle W Kimmel
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - Sven E Eklund
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - John P Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee. 37235, USA
| | - David E Cliffel
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee. 37235, USA ; Department of Chemistry, Vanderbilt University, Nashville, Tennessee. 37235, USA
| |
Collapse
|
11
|
Wikswo JP. The relevance and potential roles of microphysiological systems in biology and medicine. Exp Biol Med (Maywood) 2014; 239:1061-72. [PMID: 25187571 PMCID: PMC4330974 DOI: 10.1177/1535370214542068] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Microphysiological systems (MPS), consisting of interacting organs-on-chips or tissue-engineered, 3D organ constructs that use human cells, present an opportunity to bring new tools to biology, medicine, pharmacology, physiology, and toxicology. This issue of Experimental Biology and Medicine describes the ongoing development of MPS that can serve as in-vitro models for bone and cartilage, brain, gastrointestinal tract, lung, liver, microvasculature, reproductive tract, skeletal muscle, and skin. Related topics addressed here are the interconnection of organs-on-chips to support physiologically based pharmacokinetics and drug discovery and screening, and the microscale technologies that regulate stem cell differentiation. The initial motivation for creating MPS was to increase the speed, efficiency, and safety of pharmaceutical development and testing, paying particular regard to the fact that neither monolayer monocultures of immortal or primary cell lines nor animal studies can adequately recapitulate the dynamics of drug-organ, drug-drug, and drug-organ-organ interactions in humans. Other applications include studies of the effect of environmental toxins on humans, identification, characterization, and neutralization of chemical and biological weapons, controlled studies of the microbiome and infectious disease that cannot be conducted in humans, controlled differentiation of induced pluripotent stem cells into specific adult cellular phenotypes, and studies of the dynamics of metabolism and signaling within and between human organs. The technical challenges are being addressed by many investigators, and in the process, it seems highly likely that significant progress will be made toward providing more physiologically realistic alternatives to monolayer monocultures or whole animal studies. The effectiveness of this effort will be determined in part by how easy the constructs are to use, how well they function, how accurately they recapitulate and report human pharmacology and toxicology, whether they can be generated in large numbers to enable parallel studies, and if their use can be standardized consistent with the practices of regulatory science.
Collapse
Affiliation(s)
- John P Wikswo
- Departments of Biomedical Engineering, Molecular Physiology and Biophysics, and Physics and Astronomy, Vanderbilt University, The Vanderbilt Institute for Integrative Biosystems Research and Education, VU Station B 351807, Nashville, TN 37235-1807, USA
| |
Collapse
|
12
|
Wang Q, Schultz BD. Cholera toxin enhances Na(+) absorption across MCF10A human mammary epithelia. Am J Physiol Cell Physiol 2013; 306:C471-84. [PMID: 24371040 DOI: 10.1152/ajpcell.00181.2013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cellular mechanisms to account for the low Na(+) concentration in human milk are poorly defined. MCF10A cells, which were derived from human mammary epithelium and grown on permeable supports, exhibit amiloride- and benzamil-sensitive short-circuit current (Isc; a sensitive indicator of net ion transport), suggesting activity of the epithelial Na(+) channel ENaC. When cultured in the presence of cholera toxin (Ctx), MCF10A cells exhibit greater amiloride-sensitive Isc at all time points tested (2 h to 7 days), an effect that is not reduced with Ctx washout for 12 h. Amiloride-sensitive Isc remains elevated by Ctx in the presence of inhibitors for PKA (H-89, Rp-cAMP), PI3K (LY294002), and protein trafficking (brefeldin A). Additionally, the Ctx B subunit, alone, does not replicate these effects. RT-PCR and Western blot analyses indicate no significant increase in either the mRNA or protein expression for α-, β-, or, γ-ENaC subunits. Ctx increases the abundance of both β- and γ-ENaC in the apical membrane. Additionally, Ctx increases both phosphorylated and nonphosphorylated Nedd4-2 expression. These results demonstrate that human mammary epithelia express ENaC, which can account for the low Na(+) concentration in milk. Importantly, the results suggest that Ctx increases the expression but reduces the activity of the E3 ubiquitin ligase Nedd4-2, which would tend to reduce the ENaC retrieval and increase steady-state membrane residency. The results reveal a novel mechanism in human mammary gland epithelia by which Ctx regulates ENaC-mediated Na(+) transport, which may have inferences for epithelial ion transport regulation in other tissues throughout the body.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | | |
Collapse
|
13
|
Shinawi TF, Kimmel DW, Cliffel DE. Multianalyte microphysiometry reveals changes in cellular bioenergetics upon exposure to fluorescent dyes. Anal Chem 2013; 85:11677-80. [PMID: 24228839 PMCID: PMC3900241 DOI: 10.1021/ac402764x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fluorescent dyes have been designed for internal cellular component specificity and have been used extensively in the scientific community as a means to monitor cell growth, location, morphology, and viability. However, it is possible that the introduction of these dyes influences the basal function of the cell and, in turn, the results of these studies. Electrochemistry provides a noninvasive method for probing the unintended cellular affects of these dyes. The multianalyte microphysiometer (MAMP) is capable of simultaneous electrochemical measurement of extracellular metabolites in real-time. In this study, analytes central to cellular metabolism, glucose, lactate, oxygen, as well as extracellular acidification were monitored to determine the immediate metabolic effects of nuclear stains, including SYTO, DAPI dilactate, Hoechst 33342, and FITC dyes upon the pheochromocytoma PC-12 cells and RAW 264.7 macrophages. The experimental results revealed that the SYTO dye 13 significantly decreased glucose and oxygen consumption and increased extracellular acidification and lactate production in both cell lines, indicating a shift to anaerobic respiration. No other dyes caused significantly definitive changes in cellular metabolism upon exposure. This study shows that fluorescent dyes can have unintended effects on cellular metabolism and care should be taken when using these probes to investigate cellular function and morphology.
Collapse
Affiliation(s)
- Tesniem F. Shinawi
- Department of Chemistry, Vanderbilt University, VU Station B. Nashville, TN 37235-1822, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1809, USA
| | - Danielle W. Kimmel
- Department of Chemistry, Vanderbilt University, VU Station B. Nashville, TN 37235-1822, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1809, USA
| | - David E. Cliffel
- Department of Chemistry, Vanderbilt University, VU Station B. Nashville, TN 37235-1822, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1809, USA
| |
Collapse
|
14
|
Shotwell MS, Drake KJ, Sidorov VY, Wikswo JP. Mechanistic analysis of challenge-response experiments. Biometrics 2013; 69:741-7. [PMID: 23859366 DOI: 10.1111/biom.12066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 03/01/2013] [Accepted: 04/01/2013] [Indexed: 12/24/2022]
Abstract
We present an application of mechanistic modeling and nonlinear longitudinal regression in the context of biomedical response-to-challenge experiments, a field where these methods are underutilized. In this type of experiment, a system is studied by imposing an experimental challenge, and then observing its response. The combination of mechanistic modeling and nonlinear longitudinal regression has brought new insight, and revealed an unexpected opportunity for optimal design. Specifically, the mechanistic aspect of our approach enables the optimal design of experimental challenge characteristics (e.g., intensity, duration). This article lays some groundwork for this approach. We consider a series of experiments wherein an isolated rabbit heart is challenged with intermittent anoxia. The heart responds to the challenge onset, and recovers when the challenge ends. The mean response is modeled by a system of differential equations that describe a candidate mechanism for cardiac response to anoxia challenge. The cardiac system behaves more variably when challenged than when at rest. Hence, observations arising from this experiment exhibit complex heteroscedasticity and sharp changes in central tendency. We present evidence that an asymptotic statistical inference strategy may fail to adequately account for statistical uncertainty. Two alternative methods are critiqued qualitatively (i.e., for utility in the current context), and quantitatively using an innovative Monte-Carlo method. We conclude with a discussion of the exciting opportunities in optimal design of response-to-challenge experiments.
Collapse
Affiliation(s)
- M S Shotwell
- Vanderbilt University, Nashville, Tennessee 37232, U.S.A
| | | | | | | |
Collapse
|
15
|
Kimmel DW, Meschievitz ME, Hiatt LA, Cliffel DE. Multianalyte Microphysiometry of Macrophage Responses to Phorbol Myristate Acetate, Lipopolysaccharide, and Lipoarabinomannan. ELECTROANAL 2013; 25:1706-1712. [PMID: 25798034 PMCID: PMC4365790 DOI: 10.1002/elan.201300121] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/08/2013] [Indexed: 01/09/2023]
Abstract
This study examined the hypothesis that mycobacterial antigens generate different metabolic responses in macrophages as compared to gram-negative effectors and macrophage activators. The metabolic activation of macrophages by PMA is a useful tool for studying virulent agents and can be compared to other effectors. While phorbol myristate acetate (PMA) is commonly used to study macrophage activation, the concentration used to create this physiological response varies. The response of RAW-264.7 macrophages is concentration-dependent, where the metabolic response to high concentrations of PMA decreases suggesting deactivation. The gram-negative effector, lipopolysaccharide (LPS), was seen to promote glucose and oxygen production which were used to produce a delayed onset of oxidative burst. Pre-incubation with interferon-γ (IFN-γ) increased the effect on cell metabolism, where the synergistic effects of IFN-γ and LPS immediately initiated oxidative burst. These studies exhibited a stark contrast with lipoarabinomannan (LAM), an antigenic glycolipid component associated with the bacterial genus Mycobacterium. The presence of LAM effectively inhibits any metabolic response preventing consumption of glucose and oxygen for the promotion of oxidative burst and to ensure pathogenic proliferation. This study demonstrates for the first time the immediate inhibitory metabolic effects LAM has on macrophages, suggesting implications for future intervention studies with Mycobacterium tuberculosis.
Collapse
Affiliation(s)
| | | | | | - David E. Cliffel
- Corresponding Author: Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, VU Station B 351822, Nashville, TN 37235-1822; Phone: (615) 343-3937;
| |
Collapse
|
16
|
Wikswo JP, Block FE, Cliffel DE, Goodwin CR, Marasco CC, Markov DA, McLean DL, McLean JA, McKenzie JR, Reiserer RS, Samson PC, Schaffer DK, Seale KT, Sherrod SD. Engineering challenges for instrumenting and controlling integrated organ-on-chip systems. IEEE Trans Biomed Eng 2013; 60:682-90. [PMID: 23380852 PMCID: PMC3696887 DOI: 10.1109/tbme.2013.2244891] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The sophistication and success of recently reported microfabricated organs-on-chips and human organ constructs have made it possible to design scaled and interconnected organ systems that may significantly augment the current drug development pipeline and lead to advances in systems biology. Physiologically realistic live microHuman (μHu) and milliHuman (mHu) systems operating for weeks to months present exciting and important engineering challenges such as determining the appropriate size for each organ to ensure appropriate relative organ functional activity, achieving appropriate cell density, providing the requisite universal perfusion media, sensing the breadth of physiological responses, and maintaining stable control of the entire system, while maintaining fluid scaling that consists of ~5 mL for the mHu and ~5 μL for the μHu. We believe that successful mHu and μHu systems for drug development and systems biology will require low-volume microdevices that support chemical signaling, microfabricated pumps, valves and microformulators, automated optical microscopy, electrochemical sensors for rapid metabolic assessment, ion mobility-mass spectrometry for real-time molecular analysis, advanced bioinformatics, and machine learning algorithms for automated model inference and integrated electronic control. Toward this goal, we are building functional prototype components and are working toward top-down system integration.
Collapse
Affiliation(s)
- John P. Wikswo
- Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics, and Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Frank E. Block
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - David E. Cliffel
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | - Cody R. Goodwin
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | - Christina C. Marasco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - Dmitry A. Markov
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232-6840 USA
| | - David L. McLean
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | | | - Ronald S. Reiserer
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Philip C. Samson
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - David K. Schaffer
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Kevin T. Seale
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - Stacy D. Sherrod
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| |
Collapse
|
17
|
Kimmel DW, Dole WP, Cliffel DE. Application of multianalyte microphysiometry to characterize macrophage metabolic responses to oxidized LDL and effects of an apoA-1 mimetic. Biochem Biophys Res Commun 2013; 431:181-5. [PMID: 23313489 DOI: 10.1016/j.bbrc.2012.12.140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 12/27/2012] [Indexed: 10/27/2022]
Abstract
Although the interaction of macrophages with oxidized low density liopoprotein (oxLDL) is critical to the pathogenesis of atherosclerosis, relatively little is known about their metabolic response to oxLDL. Our development of the multianalyte microphysiometer (MAMP) allows for simultaneous measurement of extracellular metabolic substrates and products in real-time. Here, we use the MAMP to study changes in the metabolic rates of RAW-264.7 cells undergoing respiratory burst in response to oxLDL. These studies indicate that short duration exposure of macrophages to oxLDL results in time-dependent increases in glucose and oxygen consumption and in lactate production and extracellular acidification rate. Since apolipoprotein A-I (apoA-I) and apoA-I mimetics prevent experimental atherosclerosis, we hypothesized that the metabolic response of the macrophage during respiratory burst can be modulated by apoA-I mimetics. We tested this hypothesis by examining the effects of the apoA-I peptide mimetic, L-4F, alone and complexed with 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) on the macrophage metabolic response to oxLDL. L-4F and the DMPC/L-4F complexes attenuated the macrophage respiratory burst in response to oxLDL. The MAMP provides a novel approach for studying macrophage ligand-receptor interactions and cellular metabolism and our results provide new insights into the metabolic effects of oxLDL and mechanism of action of apoA-I mimetics.
Collapse
Affiliation(s)
- Danielle W Kimmel
- Department of Chemistry, Vanderbilt University, VU Station B, Nashville, TN 37235-1822, USA
| | | | | |
Collapse
|
18
|
Harry RS, Hiatt LA, Kimmel DW, Carney CK, Halfpenny KC, Cliffel DE, Wright DW. Metabolic impact of 4-hydroxynonenal on macrophage-like RAW 264.7 function and activation. Chem Res Toxicol 2012; 25:1643-51. [PMID: 22799741 DOI: 10.1021/tx3001048] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metabolic profiling of macrophage metabolic response upon exposure to 4-hydroxynonenal (HNE) demonstrates that HNE does not simply inactivate superoxide-generating enzymes but also could be responsible for the impairment of downfield signaling pathways. Multianalyte microphysiometry (MAMP) was employed to simultaneously measure perturbations in extracellular acidification, lactate production, and oxygen consumption for the examination of aerobic and anaerobic pathways. Combining the activation of oxidative burst with phorbol myristate acetate (PMA) and the immunosuppression with HNE, the complex nature of HNE toxicity was determined to be concentration- and time-dependent. Further analysis was utilized to assess the temporal effect of HNE on reactive oxygen species (ROS) production and on protein kinase C (PKC). Increased levels of HNE with decreasing PKC activity suggest that PKC is a target for HNE adductation prior to oxidative burst. Additionally, localization of PKC to the cell membrane was prevented with the introduction of HNE, demonstrating a consequence of HNE adductation on NADPH activation. The impairment of ROS by HNE suggests that HNE has a greater role in foam cell formation and tissue damage than is already known. Although work has been performed to understand the effect of HNE's regulation of specific signaling pathways, details regarding its involvement in cellular metabolism as a whole are generally unknown. This study examines the impact of HNE on macrophage oxidative burst and identifies PKC as a key protein for HNE suppression and eventual metabolic response.
Collapse
Affiliation(s)
- Reese S Harry
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, VU Station B 351822, Nashville, TN 37235-1822, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
McKenzie JR, Palubinsky AM, Brown JE, McLaughlin B, Cliffel DE. Metabolic multianalyte microphysiometry reveals extracellular acidosis is an essential mediator of neuronal preconditioning. ACS Chem Neurosci 2012; 3:510-8. [PMID: 22860220 DOI: 10.1021/cn300003r] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/22/2012] [Indexed: 01/08/2023] Open
Abstract
Metabolic adaptation to stress is a crucial yet poorly understood phenomenon, particularly in the central nervous system (CNS). The ability to identify essential metabolic events which predict neuronal fate in response to injury is critical to developing predictive markers of outcome, for interpreting CNS spectroscopic imaging, and for providing a richer understanding of the relevance of clinical indices of stress which are routinely collected. In this work, real-time multianalyte microphysiometry was used to dynamically assess multiple markers of aerobic and anaerobic respiration through simultaneous electrochemical measurement of extracellular glucose, lactate, oxygen, and acid. Pure neuronal cultures and mixed cultures of neurons and glia were compared following a 90 min exposure to aglycemia. This stress was cytotoxic to neurons yet resulted in no appreciable increase in cell death in age-matched mixed cultures. The metabolic profile of the cultures was similar in that aglycemia resulted in decreases in extracellular acidification and lactate release in both pure neurons and mixed cultures. However, oxygen consumption was only diminished in the neuron enriched cultures. The differences became more pronounced when cells were returned to glucose-containing media upon which extracellular acidification and oxygen consumption never returned to baseline in cells fated to die. Taken together, these data suggest that lactate release is not predictive of neuronal survival. Moreover, they reveal a previously unappreciated relationship of astrocytes in maintaining oxygen uptake and a correlation between metabolic recovery of neurons and extracellular acidification.
Collapse
Affiliation(s)
- Jennifer R. McKenzie
- Departments of †Chemistry, ‡Neurology, §Pharmacology, ∥Vanderbilt Brain Institute, ⊥Vanderbilt Kennedy
Center, and #Neuroscience
Graduate Program, Vanderbilt University, Nashville, Tennessee
37232, United States
| | - Amy M. Palubinsky
- Departments of †Chemistry, ‡Neurology, §Pharmacology, ∥Vanderbilt Brain Institute, ⊥Vanderbilt Kennedy
Center, and #Neuroscience
Graduate Program, Vanderbilt University, Nashville, Tennessee
37232, United States
| | - Jacquelynn E. Brown
- Departments of †Chemistry, ‡Neurology, §Pharmacology, ∥Vanderbilt Brain Institute, ⊥Vanderbilt Kennedy
Center, and #Neuroscience
Graduate Program, Vanderbilt University, Nashville, Tennessee
37232, United States
| | - BethAnn McLaughlin
- Departments of †Chemistry, ‡Neurology, §Pharmacology, ∥Vanderbilt Brain Institute, ⊥Vanderbilt Kennedy
Center, and #Neuroscience
Graduate Program, Vanderbilt University, Nashville, Tennessee
37232, United States
| | - David E. Cliffel
- Departments of †Chemistry, ‡Neurology, §Pharmacology, ∥Vanderbilt Brain Institute, ⊥Vanderbilt Kennedy
Center, and #Neuroscience
Graduate Program, Vanderbilt University, Nashville, Tennessee
37232, United States
| |
Collapse
|