1
|
Adebayo G, Ayanda OI, Rottmann M, Ajibaye OS, Oduselu G, Mulindwa J, Ajani OO, Aina O, Mäser P, Adebiyi E. The Importance of Murine Models in Determining In Vivo Pharmacokinetics, Safety, and Efficacy in Antimalarial Drug Discovery. Pharmaceuticals (Basel) 2025; 18:424. [PMID: 40143200 PMCID: PMC11944934 DOI: 10.3390/ph18030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
New chemical entities are constantly being investigated towards antimalarial drug discovery, and they require animal models for toxicity and efficacy testing. Murine models show physiological similarities to humans and are therefore indispensable in the search for novel antimalarial drugs. They provide a preclinical basis (following in vitro assessments of newly identified lead compounds) for further assessment in the drug development pipeline. Specific mouse strains, non-humanized and humanized, have successfully been infected with rodent Plasmodium species and the human Plasmodium species, respectively. Infected mice provide a platform for the assessment of treatment options being sought. In vivo pharmacokinetic evaluations are necessary when determining the fate of potential antimalarials in addition to the efficacy assessment of these chemical entities. This review describes the role of murine models in the drug development pipeline. It also explains some in vivo pharmacokinetic, safety, and efficacy parameters necessary for making appropriate choices of lead compounds in antimalarial drug discovery. Despite the advantages of murine models in antimalarial drug discovery, certain limitations are also highlighted.
Collapse
Affiliation(s)
- Glory Adebayo
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Opeyemi I. Ayanda
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Olusola S. Ajibaye
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Gbolahan Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Julius Mulindwa
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, Kampala P.O. Box 7062, Uganda;
| | - Olayinka O. Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Oluwagbemiga Aina
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- African Centre of Excellence in Bioinformatics & Data Intensive Science (ACE), Kampala P.O. Box 7062, Uganda
- Infectious Diseases Institute, Makerere University, Kampala P.O. Box 22418, Uganda
| |
Collapse
|
2
|
Shukla PK, Rao RG, Meena AS, Giorgianni F, Lee SC, Raju P, Shashikanth N, Shekhar C, Beranova S, Balazs L, Tigyi G, Gosain A, Rao R. Paneth cell dysfunction in radiation injury and radio-mitigation by human α-defensin 5. Front Immunol 2023; 14:1174140. [PMID: 37638013 PMCID: PMC10448521 DOI: 10.3389/fimmu.2023.1174140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.
Collapse
Affiliation(s)
- Pradeep K. Shukla
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Roshan G. Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Avtar S. Meena
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Francesco Giorgianni
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sue Chin Lee
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Preeti Raju
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nitesh Shashikanth
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chandra Shekhar
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sarka Beranova
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Louisa Balazs
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Gabor Tigyi
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ankush Gosain
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - RadhaKrishna Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
3
|
Giorgianni F, Beranova-Giorgianni S. Oxidized low-density lipoprotein causes ribosome reduction and inhibition of protein synthesis in retinal pigment epithelial cells. Biochem Biophys Rep 2022; 32:101345. [PMID: 36204727 PMCID: PMC9530482 DOI: 10.1016/j.bbrep.2022.101345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/27/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022] Open
Abstract
Retinal pigment epithelium (RPE) are specialized multifunctional cells indispensable for maintenance of vision. Dysfunction and death of the RPE cells is implicated in the genesis and progression of age-related macular degeneration (AMD). Oxidative stress and resulting cellular damage plays a critical mechanistic role in AMD pathogenesis. Oxidized low-density lipoprotein (oxLDL), derived from LDL in a pro-oxidative environment, is found adjacent to the RPE as part of drusen, extracellular deposits that are a characteristic clinical feature of AMD. OxLDL is cytotoxic and oxLDL-induced oxidative damage may contribute to functional impairment of the RPE. Therefore, knowledge of how the RPE respond to oxLDL exposure is important to understand the mechanisms underlying RPE dysfunction and death associated with AMD. The objective of this study was to characterize alterations in the RPE proteome triggered by exposure to non-cytotoxic levels of oxLDL. Protein identification and quantification were performed with a high -resolution LC-MS/MS-based proteomics workflow. In total, out of the ca 3000 RPE proteins quantified, oxLDL treatment caused expression changes of 303 proteins. As revealed by protein functional analysis, oxLDL uptake caused a multifaceted molecular response that involved numerous biological pathways. This response included up-regulation of anti-oxidative stress proteins whose expression is mediated by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2), confirming results of transcriptomics studies previously published by us and others. Significantly, and previously unreported, the oxLDL treatment induced down-regulation of ribosomal and translation initiation proteins, and up-regulation of proteins involved in autophagy, thus suggesting that a major cellular mechanism through which the RPE mitigate oxLDL-induced damage involves inhibition of protein synthesis and removal of misfolded proteins. OxLDL causes oxidative stress in the RPE. The proteome of the RPE is impacted by non-lethal doses of OxLDL. Differentially expressed proteins include oxidative stress response and proteins involved in protein synthesis and autophagy. Protein synthesis reduction and increase in autophagy suggest presence of misfolded proteins as a result of OxLDL exposure.
Collapse
|
4
|
Bernal V, Soancatl-Aguilar V, Bulthuis J, Guryev V, Horvatovich P, Grzegorczyk M. GeneNetTools: tests for Gaussian graphical models with shrinkage. Bioinformatics 2022; 38:5049-5054. [PMID: 36179082 PMCID: PMC9665865 DOI: 10.1093/bioinformatics/btac657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2022] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Gaussian graphical models (GGMs) are network representations of random variables (as nodes) and their partial correlations (as edges). GGMs overcome the challenges of high-dimensional data analysis by using shrinkage methodologies. Therefore, they have become useful to reconstruct gene regulatory networks from gene-expression profiles. However, it is often ignored that the partial correlations are 'shrunk' and that they cannot be compared/assessed directly. Therefore, accurate (differential) network analyses need to account for the number of variables, the sample size, and also the shrinkage value, otherwise, the analysis and its biological interpretation would turn biased. To date, there are no appropriate methods to account for these factors and address these issues. RESULTS We derive the statistical properties of the partial correlation obtained with the Ledoit-Wolf shrinkage. Our result provides a toolbox for (differential) network analyses as (i) confidence intervals, (ii) a test for zero partial correlation (null-effects) and (iii) a test to compare partial correlations. Our novel (parametric) methods account for the number of variables, the sample size and the shrinkage values. Additionally, they are computationally fast, simple to implement and require only basic statistical knowledge. Our simulations show that the novel tests perform better than DiffNetFDR-a recently published alternative-in terms of the trade-off between true and false positives. The methods are demonstrated on synthetic data and two gene-expression datasets from Escherichia coli and Mus musculus. AVAILABILITY AND IMPLEMENTATION The R package with the methods and the R script with the analysis are available in https://github.com/V-Bernal/GeneNetTools. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Victor Bernal
- Center of Information Technology, University of Groningen, Groningen 9747 AJ, The Netherlands,Department of Mathematics, Bernoulli Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| | | | - Jonas Bulthuis
- Center of Information Technology, University of Groningen, Groningen 9747 AJ, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, The Netherlands
| | | | | |
Collapse
|
5
|
TRPV6 channel mediates alcohol-induced gut barrier dysfunction and systemic response. Cell Rep 2022; 39:110937. [PMID: 35705057 PMCID: PMC9250449 DOI: 10.1016/j.celrep.2022.110937] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/20/2022] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
Intestinal epithelial tight junction disruption is a primary contributing factor in alcohol-associated endotoxemia, systemic inflammation, and multiple organ damage. Ethanol and acetaldehyde disrupt tight junctions by elevating intracellular Ca2+. Here we identify TRPV6, a Ca2+-permeable channel, as responsible for alcohol-induced elevation of intracellular Ca2+, intestinal barrier dysfunction, and systemic inflammation. Ethanol and acetaldehyde elicit TRPV6 ionic currents in Caco-2 cells. Studies in Caco-2 cell monolayers and mouse intestinal organoids show that TRPV6 deficiency or inhibition attenuates ethanol- and acetaldehyde-induced Ca2+ influx, tight junction disruption, and barrier dysfunction. Moreover, Trpv6−/− mice are resistant to alcohol-induced intestinal barrier dysfunction. Photoaffinity labeling of 3-azibutanol identifies a histidine as a potential alcohol-binding site in TRPV6. The substitution of this histidine, and a nearby arginine, reduces ethanol-activated currents. Our findings reveal that TRPV6 is required for alcohol-induced gut barrier dysfunction and inflammation. Molecules that decrease TRPV6 function have the potential to attenuate alcohol-associated tissue injury. Meena et al. show that the mechanism of alcohol-induced gut permeability, endotoxemia, and systemic inflammation requires the TRPV6 channel. They show that ethanol activates TRPV6, induces calcium influx, and disrupts intestinal epithelial tight junctions. Furthermore, specific histidine and arginine residues at the N terminus fine-tune the alcohol-induced activation of TRPV6.
Collapse
|
6
|
Guiren Fritah H, Rovelli R, Lai-Lai Chiang C, Kandalaft LE. The current clinical landscape of personalized cancer vaccines. Cancer Treat Rev 2022; 106:102383. [DOI: 10.1016/j.ctrv.2022.102383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022]
|
7
|
Long JE, Jankovic M, Maddalo D. Drug discovery oncology in a mouse: concepts, models and limitations. Future Sci OA 2021; 7:FSO737. [PMID: 34295539 PMCID: PMC8288236 DOI: 10.2144/fsoa-2021-0019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/27/2021] [Indexed: 02/08/2023] Open
Abstract
The utilization of suitable mouse models is a critical step in the drug discovery oncology workflow as their generation and use are important for target identification and validation as well as toxicity and efficacy assessments. Current murine models have been instrumental in furthering insights into the mode of action of drugs before transitioning into the clinic. Recent advancements in genome editing with the development of the CRISPR/Cas9 system and the possibility of applying such technology directly in vivo have expanded the toolkit of preclinical models available. In this review, a brief presentation of the current models used in drug discovery will be provided with a particular emphasis on the novel CRISPR/Cas9 models.
Collapse
Affiliation(s)
- Jason E Long
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Maja Jankovic
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada
- Lady Davis Institute for Medical Research, Montréal, QC, H4A 3J1, Canada
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, 4070, Switzerland
- Author for correspondence:
| |
Collapse
|
8
|
Parks C, Rogers CM, Prins P, Williams RW, Chen H, Jones BC, Moore BM, Mulligan MK. Genetic Modulation of Initial Sensitivity to Δ9-Tetrahydrocannabinol (THC) Among the BXD Family of Mice. Front Genet 2021; 12:659012. [PMID: 34367237 PMCID: PMC8343140 DOI: 10.3389/fgene.2021.659012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022] Open
Abstract
Cannabinoid receptor 1 activation by the major psychoactive component in cannabis, Δ9-tetrahydrocannabinol (THC), produces motor impairments, hypothermia, and analgesia upon acute exposure. In previous work, we demonstrated significant sex and strain differences in acute responses to THC following administration of a single dose (10 mg/kg, i.p.) in C57BL/6J (B6) and DBA/2J (D2) inbred mice. To determine the extent to which these differences are heritable, we quantified acute responses to a single dose of THC (10 mg/kg, i.p.) in males and females from 20 members of the BXD family of inbred strains derived by crossing and inbreeding B6 and D2 mice. Acute THC responses (initial sensitivity) were quantified as changes from baseline for: 1. spontaneous activity in the open field (mobility), 2. body temperature (hypothermia), and 3. tail withdrawal latency to a thermal stimulus (antinociception). Initial sensitivity to the immobilizing, hypothermic, and antinociceptive effects of THC varied substantially across the BXD family. Heritability was highest for mobility and hypothermia traits, indicating that segregating genetic variants modulate initial sensitivity to THC. We identified genomic loci and candidate genes, including Ndufs2, Scp2, Rps6kb1 or P70S6K, Pde4d, and Pten, that may control variation in THC initial sensitivity. We also detected strong correlations between initial responses to THC and legacy phenotypes related to intake or response to other drugs of abuse (cocaine, ethanol, and morphine). Our study demonstrates the feasibility of mapping genes and variants modulating THC responses in the BXDs to systematically define biological processes and liabilities associated with drug use and abuse.
Collapse
Affiliation(s)
- Cory Parks
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Agriculture, Biology and Health Sciences, Cameron University, Lawton, OK, United States
| | - Chris M. Rogers
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Pjotr Prins
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C. Jones
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Bob M. Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
9
|
Kumagai A, Takeda S, Sohara E, Uchida S, Iijima H, Itakura A, Koya D, Kanasaki K. Dietary Magnesium Insufficiency Induces Salt-Sensitive Hypertension in Mice Associated With Reduced Kidney Catechol-O-Methyl Transferase Activity. Hypertension 2021; 78:138-150. [PMID: 33840199 DOI: 10.1161/hypertensionaha.120.16377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Asako Kumagai
- Faculty of Medicine, Internal Medicine I, Shimane University, Izumo, Japan (A.K., K.K.)
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Juntendo University, Bunkyo, Tokyo, Japan (A.K., S.T., A.I.)
- Department of Diabetology and Endocrinology (A.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Juntendo University, Bunkyo, Tokyo, Japan (A.K., S.T., A.I.)
| | - Eisei Sohara
- Department of Nephrology, Tokyo Medical and Dental University, Yushima, Bunkyo, Japan (E.S., S.U.)
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University, Yushima, Bunkyo, Japan (E.S., S.U.)
| | - Hiroshi Iijima
- School of Pharmacy, Nihon University, Chiba, Japan (H.I.)
| | - Astuo Itakura
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Juntendo University, Bunkyo, Tokyo, Japan (A.K., S.T., A.I.)
| | - Daisuke Koya
- Department of Diabetology and Endocrinology (A.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (D.K., K.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Keizo Kanasaki
- Faculty of Medicine, Internal Medicine I, Shimane University, Izumo, Japan (A.K., K.K.)
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (D.K., K.K.), Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
10
|
Parks C, Jones BC, Moore BM, Mulligan MK. Sex and Strain Variation in Initial Sensitivity and Rapid Tolerance to Δ9-Tetrahydrocannabinol. Cannabis Cannabinoid Res 2020; 5:231-245. [PMID: 32923660 PMCID: PMC7480727 DOI: 10.1089/can.2019.0047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background and Objectives: For cannabis and other drugs of abuse, initial response and/or tolerance to drug effects can predict later dependence and problematic use. Our objective is to identify sex and genetic (strain) differences in initial response and rapid tolerance to Δ9–tetrahydrocannabinol (THC), the main psychoactive ingredient in cannabis, between highly genetically divergent inbred mouse strains—C57BL/6J (B6) and DBA/2J (D2). Experimental Approach: Sex and strain responses relative to baseline were quantified following daily exposure (i.p.) to 10 mg/kg THC or vehicle (VEH) over the course of 5 days. Dependent measures included hypothermia (decreased body temperature) and ataxia (decreased spontaneous activity in the open field), and antinociception (increase in tail withdrawal latency to a thermal stimulus). Initial sensitivity to THC was defined as the difference in response between baseline and day 1. Rapid tolerance to THC was defined as the difference in response between days 1 and 2. Results: B6 exhibited greater THC-induced motor activity suppression and initial sensitivity to ataxia relative to the D2 strain. Females demonstrated greater levels of THC-induced hypothermia and initial sensitivity relative to males. Higher levels of THC-induced antinociception and initial sensitivity were observed for D2 relative to B6. Rapid tolerance to THC was observed for hypothermia and antinociception. Much less tolerance was observed for THC-induced ataxia. D2 exhibited rapid tolerance to THC-induced hypothermia and antinociception at time points associated with peak THC initial response. Likewise, at the peak initial THC response time point, females demonstrated greater levels of rapid tolerance to hypothermic effects relative to males. Conclusions: Both sex and genetic factors drive variation in initial response and rapid tolerance to the ataxic, antinociceptive, and hypothermic effects of THC. As these traits directly result from THC activation of the cannabinoid receptor 1, gene variants between B6 and D2 in cannabinoid signaling pathways are likely to mediate strain differences in response to THC.
Collapse
Affiliation(s)
- Cory Parks
- Department of Genetics, Genomics and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Byron C Jones
- Department of Genetics, Genomics and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Bob M Moore
- Department of Pharmaceutical Sciences, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
11
|
De Majo F, Hegenbarth JC, Rühle F, Bär C, Thum T, de Boer M, Duncker DJ, Schroen B, Armand AS, Stoll M, De Windt LJ. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts. Sci Rep 2020; 10:8136. [PMID: 32424227 PMCID: PMC7235007 DOI: 10.1038/s41598-020-65115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/29/2020] [Indexed: 11/12/2022] Open
Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
Collapse
Affiliation(s)
- Federica De Majo
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Frank Rühle
- Bioinformatics Core Facility, Institute of Molecular Biology (IMB), Mainz, Germany.,Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Martine de Boer
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Blanche Schroen
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Anne-Sophie Armand
- Institut Necker Enfants Malades, Inserm U1151, Paris, France; Universite Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Leon J De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands. .,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|