1
|
Lu EH, Rusyn I, Chiu WA. Incorporating new approach methods (NAMs) data in dose-response assessments: The future is now! JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025; 28:28-62. [PMID: 39390665 PMCID: PMC11614695 DOI: 10.1080/10937404.2024.2412571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Regulatory dose-response assessments traditionally rely on in vivo data and default assumptions. New Approach Methods (NAMs) present considerable opportunities to both augment traditional dose-response assessments and accelerate the evaluation of new/data-poor chemicals. This review aimed to determine the potential utilization of NAMs through a unified conceptual framework that compartmentalizes derivation of toxicity values into five sequential Key Dose-response Modules (KDMs): (1) point-of-departure (POD) determination, (2) test system-to-human (e.g. inter-species) toxicokinetics and (3) toxicodynamics, (4) human population (intra-species) variability in toxicodynamics, and (5) toxicokinetics. After using several "traditional" dose-response assessments to illustrate this framework, a review is presented where existing NAMs, including in silico, in vitro, and in vivo approaches, might be applied across KDMs. Further, the false dichotomy between "traditional" and NAMs-derived data sources is broken down by organizing dose-response assessments into a matrix where each KDM has Tiers of increasing precision and confidence: Tier 0: Default/generic values, Tier 1: Computational predictions, Tier 2: Surrogate measurements, and Tier 3: Direct measurements. These findings demonstrated that although many publications promote the use of NAMs in KDMs (1) for POD determination and (5) for human population toxicokinetics, the proposed matrix of KDMs and Tiers reveals additional immediate opportunities for NAMs to be integrated across other KDMs. Further, critical needs were identified for developing NAMs to improve in vitro dosimetry and quantify test system and human population toxicodynamics. Overall, broadening the integration of NAMs across the steps of dose-response assessment promises to yield higher throughput, less animal-dependent, and more science-based toxicity values for protecting human health.
Collapse
Affiliation(s)
- En-Hsuan Lu
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States of America
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States of America
| | - Weihsueh A. Chiu
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, United States of America
| |
Collapse
|
2
|
Sweet MG, Iglesias-Carres L, Ellsworth PN, Carter JD, Nielsen DM, Aylor DL, Tessem JS, Neilson AP. Phenotype variability in diet-induced obesity and response to (-)-epigallocatechin gallate supplementation in a Diversity Outbred mouse cohort: A model for exploring gene x diet interactions for dietary bioactives. Nutr Res 2025; 133:78-93. [PMID: 39705912 DOI: 10.1016/j.nutres.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
The flavan-3-ol (-)-epigallocatechin gallate (EGCG) blunts obesity in inbred mice, but human clinical trials have yielded mixed results. Genetic homogeneity in preclinical models may explain translational disconnect between rodents and humans. The Diversity Outbred (DO) mouse model provides genotype and phenotype variability for characterization of gene x environment (i.e., diet) interactions. We conducted a longitudinal phenotyping study in DO mice. Mice (n = 50) were fed a high-fat diet for 8 weeks and then a high-fat diet + 0.3% EGCG for 8 weeks. We hypothesized that obesity and any protective effects of EGCG would exhibit extreme variability in these genetically heterogeneous mice. As anticipated, DO mice exhibited extreme variation in body composition at baseline (4%-13.9% fat), after 8 weeks of high-fat diet (6.5%-38.1% fat), and after 8 weeks of high-fat diet + EGCG (7.6%-42.6% fat), greater than what is observed in inbred mice. All 50 mice gained body fat on the high-fat diet (changes from baseline of +5% ± 640%). Intriguingly, adiposity variability increased when EGCG was added to the diet (changes from the high-fat diet alone of -52% ± 390%), with 11/50 mice losing body fat. We postulate that the explanation for this variability is genetic heterogeneity. Our data confirm the promise for EGCG to manage obesity but suggest that genetic factors may exert significant control over the efficacy of EGCG. Larger studies in DO mice are needed for quantitative trait loci mapping to identify genetic loci governing EGCG x obesity interactions and translate these findings to precision nutrition in humans.
Collapse
Affiliation(s)
- Michael G Sweet
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | | | - Peter N Ellsworth
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Jared D Carter
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Dahlia M Nielsen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - David L Aylor
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Jeffery S Tessem
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Andrew P Neilson
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA; Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
3
|
Hong S, Nguyen BN, Min H, Youn HY, Choi S, Hitayezu E, Cha KH, Park YT, Lee CG, Yoo G, Kim M. Host-specific effects of Eubacterium species on Rg3-mediated modulation of osteosarcopenia in a genetically diverse mouse population. MICROBIOME 2024; 12:251. [PMID: 39623488 PMCID: PMC11613481 DOI: 10.1186/s40168-024-01971-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/08/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Osteosarcopenia, characterized by the simultaneous loss of bone and muscle mass, is a serious health problem in the aging population. This study investigated the interplay between host genetics, gut microbiota, and musculoskeletal health in a mouse model of osteosarcopenia, exploring the therapeutic potential of gut microbiota modulation. METHODS We examined the effects of Rg3, a phytochemical, on osteosarcopenia and its interactions with host genetics and gut microbiota in six founder strains of the Collaborative Cross (CC) population. Subsequently, we evaluated the therapeutic potential of Eubacterium nodatum (EN) and Eubacterium ventriosum (EV), two gut microbes identified as significant correlates of Rg3-mediated osteosarcopenia improvement, in selected C57BL/6 J (B6) and 129S1/SvImJ (129S1) mouse strains. RESULTS Rg3 treatment altered gut microbiota composition aligned with osteosarcopenia phenotypes, which response varied depending on host genetics. This finding enabled the identification of two microbes in the Eubacterium genus, potential mediator of Rg3 effect on osteosarcopenia. Oral administration of EN and EV differentially impacted bone density, muscle mass, exercise performance, and related gene expression in a mouse strain-specific manner. In 129S1 mice, EN and EV significantly improved these parameters, effectively reversing osteosarcopenic phenotypes. Mechanistic investigations revealed that these effects were mediated through the modulation of osteoblast differentiation and protein degradation pathways. In contrast, EN and EV did not significantly improve osteosarcopenic phenotypes in B6 mice, although they did modulate mitochondrial biogenesis and microbial diversity. CONCLUSIONS Our findings underscore the complex interplay between host genetics and the gut microbiota in osteosarcopenia and emphasize the need for personalized treatment strategies. EN and EV exhibit strain-specific therapeutic effects, suggesting that tailoring microbial interventions to individual genetic backgrounds may be crucial for optimizing treatment outcomes. Video Abstract.
Collapse
Affiliation(s)
- Soyeon Hong
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, Gangneung, Gangwon-Do, 25451, Republic of Korea
| | - Bao Ngoc Nguyen
- College of Dentistry, Gangneung Wonju National University, Gangneung, Gangwon-Do, Republic of Korea
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea
| | - Huitae Min
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea
| | - Hye-Young Youn
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea
| | - Sowoon Choi
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea
| | - Emmanuel Hitayezu
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology (KIST) Gangneung Institute, Gangneung, 25451, Republic of Korea
| | - Kwang-Hyun Cha
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology (KIST) Gangneung Institute, Gangneung, 25451, Republic of Korea
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-Do, Republic of Korea
| | - Young Tae Park
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Choong-Gu Lee
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology (KIST) Gangneung Institute, Gangneung, 25451, Republic of Korea
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-Do, Republic of Korea
| | - GyHye Yoo
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, Gangneung, Gangwon-Do, 25451, Republic of Korea.
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | - Myungsuk Kim
- Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, 679 Saimdang-Ro, Gangneung, Gangwon-Do, 210-340, Republic of Korea.
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-Do, Republic of Korea.
| |
Collapse
|
4
|
Kiaris H. Nontraditional models as research tools: the road not taken. Trends Mol Med 2024; 30:924-931. [PMID: 39069395 PMCID: PMC11466687 DOI: 10.1016/j.molmed.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Historical reasons resulted in the almost exclusive use of a few species, most prominently Mus musculus, as the mainstream models in biomedical research. This selection was not based on Mus's distinctive relevance to human disease but rather to the pre-existing availability of resources and tools for the species that were used as models, which has enabled their adoption for research in health sciences. Unless the utilization and range of nontraditional research models expand considerably, progress in biomedical research will remain restricted within the trajectory that has been set by the existing models and their ability to provide clinically relevant information.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy and Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
5
|
Getsy PM, Coffee GA, Bates JN, Parran T, Hoffer L, Baby SM, MacFarlane PM, Knauss ZT, Damron DS, Hsieh YH, Bubier JA, Mueller D, Lewis SJ. The cell-permeant antioxidant D-thiol ester D-cysteine ethyl ester overcomes physical dependence to morphine in male Sprague Dawley rats. Front Pharmacol 2024; 15:1444574. [PMID: 39253377 PMCID: PMC11381264 DOI: 10.3389/fphar.2024.1444574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The ability of morphine to decrease cysteine transport into neurons by inhibition of excitatory amino acid transporter 3 (EAA3) may be a key molecular mechanism underlying the acquisition of physical and psychological dependence to morphine. This study examined whether co-administration of the cell-penetrant antioxidant D-thiol ester, D-cysteine ethyl ester (D-CYSee), with morphine, would diminish the development of physical dependence to morphine in male Sprague Dawley rats. Systemic administration of the opioid receptor antagonist, naloxone (NLX), elicited pronounced withdrawal signs (e.g., wet-dog shakes, jumps, rears, circling) in rats that received a subcutaneous depot of morphine (150 mg/kg, SC) for 36 h and continuous intravenous infusion of vehicle (20 μL/h, IV). The NLX-precipitated withdrawal signs were reduced in rats that received an infusion of D-CYSee, but not D-cysteine, (both at 20.8 μmol/kg/h, IV) for the full 36 h. NLX elicited pronounced withdrawal signs in rats treated for 48 h with morphine (150 mg/kg, SC), plus continuous infusion of vehicle (20 μL/h, IV) that began at the 36 h timepoint of morphine treatment. The NLX-precipitated withdrawal signs were reduced in rats that received a 12 h infusion of D-CYSee, but not D-cysteine, (both at 20.8 μmol/kg/h, IV) that began at the 36 h timepoint of morphine treatment. These findings suggest that D-CYSee may attenuate the development of physical dependence to morphine and reverse established dependence to the opioid in male Sprague Dawley rats. Alternatively, D-CYSee may simply suppress the processes responsible for NLX-precipitated withdrawal. Nonetheless, D-CYSee and analogues may be novel therapeutics for the treatment of opioid use disorders.
Collapse
Affiliation(s)
- Paulina M. Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Gregory A. Coffee
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - James N. Bates
- Department of Anesthesiology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Theodore Parran
- Center for Medical Education, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Lee Hoffer
- Department of Anthropology, Case Western Reserve University, Cleveland, OH, United States
| | - Santhosh M. Baby
- Section of Biology, Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | - Peter M. MacFarlane
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Zackery T. Knauss
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Derek S. Damron
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, United States
| | | | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
- Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
6
|
Rahmah Z, Nirmala KC, Nashichuddin A, Riskiyana R, Milliana A, Indriana N, Astari LF, Ayudianti P, Kholil M. Effect of neem leaf extract ( Azadirachta indica) in reducing the degree of parasitemia and apoptosis in C57BL mice with cerebral malaria. Vet World 2024; 17:1497-1503. [PMID: 39185035 PMCID: PMC11344125 DOI: 10.14202/vetworld.2024.1497-1503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/07/2024] [Indexed: 08/27/2024] Open
Abstract
Background and Aim Brain malaria, which results from Plasmodium falciparum infection, is responsible for substantial fatalities and health issues. These processes, including cytoadherence, rosetting, and sequestration, induce an immune response, hypoxia, brain microvascular obstruction, disruption of the blood-brain barrier, and cell death. Parasitemia level can reveal the presence of infection and its association with apoptosis-related genes. Neem (Azadirachta indica) leaves with antimalarial properties could replace ineffective Indonesian malaria medications. This study was designed to evaluate the impact of neem leaf extract on cerebral malaria-induced parasitemia and neuron cell apoptosis in mice through an in vivo approach. Materials and Methods 13-16 weeks old C57BL mice received infection by Plasmodium berghei strain ANKA. Parasitemia was estimated daily from the mice's tail blood. 8 mg, 12 mg, and 16 mg of a 96% ethanolic neem leaf extract were orally given for 6 days. Healthy, positive, and negative controls were included for treatment comparisons. On the 7th day, brain tissue was analyzed for (p > 0.05) gene expression. Through immunohistochemistry, both cell apoptosis in neurons expressing caspase-3 within a brain sample and the degree of parasitemia in a blood smear were assessed. The Pearson correlation test and one-way analysis of variance were employed to analyze the data. Results Neem leaf extract reduces parasitemia and neuron cell apoptosis at multiple dosages (p < 0.000). Apoptosis in brain neurons and parasitemia show a strong positive correlation (r = +0.939). Neem leaf extract at doses of 12 and 16 mg was the most effective in reducing parasitemia levels and causing cell death. Conclusions Neem leaf therapy significantly reduced the degree of parasitemia and cell apoptosis in C57BL mice compared with the control group without treatment (p = 0.05). This shows that neem leaves have the potential to be a candidate drug for malaria.
Collapse
Affiliation(s)
- Zainabur Rahmah
- Department of Parasitology, Faculty of Medicine and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Kautsar Citra Nirmala
- Medicine Study Program, Faculty of Medicine and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Ach Nashichuddin
- Department of Mathematics, Faculty of Science and Technology, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Riskiyana Riskiyana
- Department of Neurology, Faculty of Medicine and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Alvi Milliana
- Department of Microbiology, Faculty of Medicine, and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Nurfianti Indriana
- Department of Obstetrics and Gynecology, Faculty of Medicine, and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Lina Fitria Astari
- Department of Child Health, Faculty of Medicine, and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Prida Ayudianti
- Department of Dermatology and Venereology, Faculty of Medicine, and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim Malang, Indonesia
| | - Munawar Kholil
- Department of Agriculture Product Technology, Politeknik Negeri Ketapang, Ketapang, Indonesia
| |
Collapse
|
7
|
Bates JN, Getsy PM, Coffee GA, Baby SM, MacFarlane PM, Hsieh YH, Knauss ZT, Bubier JA, Mueller D, Lewis SJ. Lipophilic analogues of D-cysteine prevent and reverse physical dependence to fentanyl in male rats. Front Pharmacol 2024; 14:1336440. [PMID: 38645835 PMCID: PMC11026688 DOI: 10.3389/fphar.2023.1336440] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/31/2023] [Indexed: 04/23/2024] Open
Abstract
We examined whether co-injections of the cell-permeant D-cysteine analogues, D-cysteine ethyl ester (D-CYSee) and D-cysteine ethyl amide (D-CYSea), prevent acquisition of physical dependence induced by twice-daily injections of fentanyl, and reverse acquired dependence to these injections in freely-moving male Sprague Dawley rats. Injection of the opioid receptor antagonist, naloxone HCl (NLX, 1.5 mg/kg, IV), elicited a series of withdrawal phenomena that included cardiorespiratory and behavioral responses, and falls in body weight and body temperature, in rats that received 5 or 10 injections of fentanyl (125 μg/kg, IV), and the same number of vehicle co-injections. Regarding the development of physical dependence, the NLX-precipitated withdrawal phenomena were markedly reduced in fentanyl-injected rats that had received co-injections of D-CYSee (250 μmol/kg, IV) or D-CYSea (100 μmol/kg, IV), but not D-cysteine (250 μmol/kg, IV). Regarding reversal of established dependence to fentanyl, the NLX-precipitated withdrawal phenomena in rats that had received 10 injections of fentanyl (125 μg/kg, IV) was markedly reduced in rats that received co-injections of D-CYSee (250 μmol/kg, IV) or D-CYSea (100 μmol/kg, IV), but not D-cysteine (250 μmol/kg, IV), starting with injection 6 of fentanyl. This study provides evidence that co-injections of D-CYSee and D-CYSea prevent the acquisition of physical dependence, and reverse acquired dependence to fentanyl in male rats. The lack of effect of D-cysteine suggests that the enhanced cell-penetrability of D-CYSee and D-CYSea into cells, particularly within the brain, is key to their ability to interact with intracellular signaling events involved in acquisition to physical dependence to fentanyl.
Collapse
Affiliation(s)
- James N. Bates
- Department of Anesthesiology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Paulina M. Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Gregory A. Coffee
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Santhosh M. Baby
- Section of Biology, Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | - Peter M. MacFarlane
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Zackery T. Knauss
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | | | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
- Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
8
|
Iglesias-Carres L, Neilson AP. Utilizing preclinical models of genetic diversity to improve translation of phytochemical activities from rodents to humans and inform personalized nutrition. Food Funct 2021; 12:11077-11105. [PMID: 34672309 DOI: 10.1039/d1fo02782d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse models are an essential tool in different areas of research, including nutrition and phytochemical research. Traditional inbred mouse models have allowed the discovery of therapeutical targets and mechanisms of action and expanded our knowledge of health and disease. However, these models lack the genetic variability typically found in human populations, which hinders the translatability of the results found in mice to humans. The development of genetically diverse mouse models, such as the collaborative cross (CC) or the diversity outbred (DO) models, has been a useful tool to overcome this obstacle in many fields, such as cancer, immunology and toxicology. However, these tools have not yet been widely adopted in the field of phytochemical research. As demonstrated in other disciplines, use of CC and DO models has the potential to provide invaluable insights for translation of phytochemicals from rodents to humans, which are desperately needed given the challenges and numerous failed clinical trials in this field. These models may prove informative for personalized use of phytochemicals in humans, including: predicting interindividual variability in phytochemical bioavailability and efficacy, identifying genetic loci or genes governing response to phytochemicals, identifying phytochemical mechanisms of action and therapeutic targets, and understanding the impact of genetic variability on individual response to phytochemicals. Such insights would prove invaluable for personalized implementation of phytochemicals in humans. This review will focus on the current work performed with genetically diverse mouse populations, and the research opportunities and advantages that these models can offer to phytochemical research.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| |
Collapse
|
9
|
Mosedale M, Cai Y, Eaddy JS, Kirby PJ, Wolenski FS, Dragan Y, Valdar W. Human-relevant mechanisms and risk factors for TAK-875-Induced liver injury identified via a gene pathway-based approach in Collaborative Cross mice. Toxicology 2021; 461:152902. [PMID: 34418498 PMCID: PMC8936092 DOI: 10.1016/j.tox.2021.152902] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Development of TAK-875 was discontinued when a small number of serious drug-induced liver injury (DILI) cases were observed in Phase 3 clinical trials. Subsequent studies have identified hepatocellular oxidative stress, mitochondrial dysfunction, altered bile acid homeostasis, and immune response as mechanisms of TAK-875 DILI and the contribution of genetic risk factors in oxidative response and mitochondrial pathways to the toxicity susceptibility observed in patients. We tested the hypothesis that a novel preclinical approach based on gene pathway analysis in the livers of Collaborative Cross mice could be used to identify human-relevant mechanisms of toxicity and genetic risk factors at the level of the hepatocyte as reported in a human genome-wide association study. Eight (8) male mice (4 matched pairs) from each of 45 Collaborative Cross lines were treated with a single oral (gavage) dose of either vehicle or 600 mg/kg TAK-875. As expected, liver injury was not detected histologically and few changes in plasma biomarkers of hepatotoxicity were observed. However, gene expression profiling in the liver identified hundreds of transcripts responsive to TAK-875 treatment across all strains reflecting alterations in immune response and bile acid homeostasis and the interaction of treatment and strain reflecting oxidative stress and mitochondrial dysfunction. Fold-change expression values were then used to develop pathway-based phenotypes for genetic mapping which identified candidate risk factor genes for TAK-875 toxicity susceptibility at the level of the hepatocyte. Taken together, these findings support our hypothesis that a gene pathway-based approach using Collaborative Cross mice could inform sensitive strains, human-relevant mechanisms of toxicity, and genetic risk factors for TAK-875 DILI. This novel preclinical approach may be helpful in understanding, predicting, and ultimately preventing clinical DILI for other drugs.
Collapse
Affiliation(s)
- Merrie Mosedale
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC, 27599, United States.
| | - Yanwei Cai
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| | - J Scott Eaddy
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC, 27599, United States.
| | - Patrick J Kirby
- Takeda Pharmaceuticals International Co., Cambridge, MA, 02139, United States.
| | - Francis S Wolenski
- Takeda Pharmaceuticals International Co., Cambridge, MA, 02139, United States.
| | - Yvonne Dragan
- Takeda Pharmaceuticals International Co., Cambridge, MA, 02139, United States.
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
10
|
Mosedale M, Watkins PB. Understanding Idiosyncratic Toxicity: Lessons Learned from Drug-Induced Liver Injury. J Med Chem 2020; 63:6436-6461. [PMID: 32037821 DOI: 10.1021/acs.jmedchem.9b01297] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Idiosyncratic adverse drug reactions (IADRs) encompass a diverse group of toxicities that can vary by drug and patient. The complex and unpredictable nature of IADRs combined with the fact that they are rare makes them particularly difficult to predict, diagnose, and treat. Common clinical characteristics, the identification of human leukocyte antigen risk alleles, and drug-induced proliferation of lymphocytes isolated from patients support a role for the adaptive immune system in the pathogenesis of IADRs. Significant evidence also suggests a requirement for direct, drug-induced stress, neoantigen formation, and stimulation of an innate response, which can be influenced by properties intrinsic to both the drug and the patient. This Perspective will provide an overview of the clinical profile, mechanisms, and risk factors underlying IADRs as well as new approaches to study these reactions, focusing on idiosyncratic drug-induced liver injury.
Collapse
Affiliation(s)
- Merrie Mosedale
- Institute for Drug Safety Sciences and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Paul B Watkins
- Institute for Drug Safety Sciences and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
11
|
Hermans E, Hulleman E. Patient-Derived Orthotopic Xenograft Models of Pediatric Brain Tumors: In a Mature Phase or Still in Its Infancy? Front Oncol 2020; 9:1418. [PMID: 31970083 PMCID: PMC6960099 DOI: 10.3389/fonc.2019.01418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
In recent years, molecular profiling has led to the discovery of an increasing number of brain tumor subtypes, and associated therapeutic targets. These molecular features have been incorporated in the 2016 new World Health Organization (WHO) Classification of Tumors of the Central Nervous System (CNS), which now distinguishes tumor subgroups not only histologically, but also based on molecular characteristics. Despite an improved diagnosis of (pediatric) tumors in the CNS however, the survival of children with malignant brain tumors still is far worse than for those suffering from other types of malignancies. Therefore, new treatments need to be developed, based on subgroup-specific genetic aberrations. Here, we provide an overview of the currently available orthotopic xenograft models for pediatric brain tumor subtypes as defined by the 2016 WHO classification, to facilitate the choice of appropriate animal models for the preclinical testing of novel treatment strategies, and to provide insight into the current gaps and challenges.
Collapse
Affiliation(s)
- Eva Hermans
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
Sawant-Basak A, Obach RS. Emerging Models of Drug Metabolism, Transporters, and Toxicity. Drug Metab Dispos 2019; 46:1556-1561. [PMID: 30333205 DOI: 10.1124/dmd.118.084293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022] Open
Abstract
This commentary summarizes expert mini-reviews and original research articles that have been assembled in a special issue on novel models of drug metabolism and disposition. The special issue consists of research articles or reviews on novel static or micro-flow based models of the intestine, liver, eye, and kidney. This issue reviews static intestinal systems like mucosal scrapings and cryopreserved intestinal enterocytes, as well as novel bioengineered or chemically engineered intestinal models derived from primary human tissue, iPSCs, enteroids, and crypts. Experts have reviewed hepatic systems like cryopermeabilized Metmax hepatocytes and longer term, hepatocyte coculture system from HµREL, yielding in vivo-like primary and secondary drug metabolite profiles. Additional liver models, including micropattern hepatocyte coculture, 3D liver spheroids, and microflow systems, applicable to the study of drug disposition and toxicology have also been reviewed. In this commentary, we have outlined expert opinions and current efforts on hepatic- and nephrotoxicity models. Ocular disposition models including corneal permeability models have been included within this special issue. This commentary provides a summary of in vivo mini-reviews of the issue, which have discussed the applications and drawbacks of pig and humanized mice models of P450, UGT, and rat organic anionic transporting polypeptide 1a4. While not extensively reviewed, novel positron emissions tomography imaging-based approaches to study the distribution of xenobiotics have been highlighted. This commentary also outlines in vitro and in vivo models of drug metabolism derived from breakthrough genetic, chromosomal, and tissue engineering techniques. The commentary concludes by providing a futuristic view of the novel models discussed in this issue.
Collapse
Affiliation(s)
- Aarti Sawant-Basak
- Pfizer Worldwide Research & Development, Clinical Pharmacology, Cambridge, Massachusetts (A.S.-B.) and Pfizer Worldwide Research & Development, Pharmacokinetics, Dynamics, and Metabolism, Groton, Connecticut (R.S.O.)
| | - R Scott Obach
- Pfizer Worldwide Research & Development, Clinical Pharmacology, Cambridge, Massachusetts (A.S.-B.) and Pfizer Worldwide Research & Development, Pharmacokinetics, Dynamics, and Metabolism, Groton, Connecticut (R.S.O.)
| |
Collapse
|
13
|
Cook JC, Wu H, Aleo MD, Adkins K. Principles of precision medicine and its application in toxicology. J Toxicol Sci 2018; 43:565-577. [PMID: 30298845 DOI: 10.2131/jts.43.565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Precision medicine is an approach to developing drugs that focuses on employing biomarkers to stratify patients in clinical trials with the goal of improving efficacy and/or safety outcomes, ultimately increasing the odds of clinical success and drug approval. Precision medicine is an important tool for toxicologists to utilize, because its principles can be used to decide whether to pursue a drug target, to understand interindividual differences in response to drugs in both nonclinical and clinical settings, to aid in selecting doses that optimize efficacy or reduce adverse events, and to facilitate understanding of a drug's mode-of-action. Nonclinical models such as the mouse and non-human primate can be used to understand genetic variation and its potential translation to humans, and are available for toxicologists to employ in advance of drugs moving into clinical development. Understanding interindividual differences in response to drugs and how these differences can influence the drug's risk-benefit profile and lead to the identification of biomarkers that enhance patient efficacy and safety is of critical importance for toxicologists today, and in the future, as the fields of pharmacogenomics and genetics continue to advance.
Collapse
Affiliation(s)
- Jon C Cook
- Pfizer Worldwide Research and Development, Groton, CT 06340
| | - Hong Wu
- Pfizer Worldwide Research and Development, Groton, CT 06340
| | - Michael D Aleo
- Pfizer Worldwide Research and Development, Groton, CT 06340
| | - Karissa Adkins
- Pfizer Worldwide Research and Development, Groton, CT 06340
| |
Collapse
|