1
|
Jaume G, de Brot S, Song AH, Williamson DFK, Oldenburg L, Zhang A, Chen RJ, Asin J, Blatter S, Dettwiler M, Goepfert C, Grau-Roma L, Soto S, Keller SM, Rottenberg S, del-Pozo J, Pettit R, Le LP, Mahmood F. Deep Learning-based Modeling for Preclinical Drug Safety Assessment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604430. [PMID: 39091793 PMCID: PMC11291027 DOI: 10.1101/2024.07.20.604430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In drug development, assessing the toxicity of candidate compounds is crucial for successfully transitioning from preclinical research to early-stage clinical trials. Drug safety is typically assessed using animal models with a manual histopathological examination of tissue sections to characterize the dose-response relationship of the compound - a time-intensive process prone to inter-observer variability and predominantly involving tedious review of cases without abnormalities. Artificial intelligence (AI) methods in pathology hold promise to accelerate this assessment and enhance reproducibility and objectivity. Here, we introduce TRACE, a model designed for toxicologic liver histopathology assessment capable of tackling a range of diagnostic tasks across multiple scales, including situations where labeled data is limited. TRACE was trained on 15 million histopathology images extracted from 46,734 digitized tissue sections from 157 preclinical studies conducted on Rattus norvegicus. We show that TRACE can perform various downstream toxicology tasks spanning histopathological response assessment, lesion severity scoring, morphological retrieval, and automatic dose-response characterization. In an independent reader study, TRACE was evaluated alongside ten board-certified veterinary pathologists and achieved higher concordance with the consensus opinion than the average of the pathologists. Our study represents a substantial leap over existing computational models in toxicology by offering the first framework for accelerating and automating toxicological pathology assessment, promoting significant progress with faster, more consistent, and reliable diagnostic processes.
Collapse
Affiliation(s)
- Guillaume Jaume
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Simone de Brot
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
| | - Andrew H. Song
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Drew F. K. Williamson
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lukas Oldenburg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Andrew Zhang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Richard J. Chen
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Javier Asin
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino, CA
- School of Veterinary Medicine, Department of Pathology, University of California-Davis, Davis, CA
| | - Sohvi Blatter
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
| | | | - Christine Goepfert
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
| | - Llorenç Grau-Roma
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
| | - Sara Soto
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
| | | | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
- Department for BioMedical Research, University of Bern, Switzerland
| | - Jorge del-Pozo
- Royal (Dick) School of Veterinary Studies, Roslin, United-Kingdom
| | - Rowland Pettit
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| |
Collapse
|
2
|
Kaplan BLF, Hoberman AM, Slikker W, Smith MA, Corsini E, Knudsen TB, Marty MS, Sobrian SK, Fitzpatrick SC, Ratner MH, Mendrick DL. Protecting Human and Animal Health: The Road from Animal Models to New Approach Methods. Pharmacol Rev 2024; 76:251-266. [PMID: 38351072 PMCID: PMC10877708 DOI: 10.1124/pharmrev.123.000967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/18/2023] [Accepted: 12/01/2023] [Indexed: 02/16/2024] Open
Abstract
Animals and animal models have been invaluable for our current understanding of human and animal biology, including physiology, pharmacology, biochemistry, and disease pathology. However, there are increasing concerns with continued use of animals in basic biomedical, pharmacological, and regulatory research to provide safety assessments for drugs and chemicals. There are concerns that animals do not provide sufficient information on toxicity and/or efficacy to protect the target population, so scientists are utilizing the principles of replacement, reduction, and refinement (the 3Rs) and increasing the development and application of new approach methods (NAMs). NAMs are any technology, methodology, approach, or assay used to understand the effects and mechanisms of drugs or chemicals, with specific focus on applying the 3Rs. Although progress has been made in several areas with NAMs, complete replacement of animal models with NAMs is not yet attainable. The road to NAMs requires additional development, increased use, and, for regulatory decision making, usually formal validation. Moreover, it is likely that replacement of animal models with NAMs will require multiple assays to ensure sufficient biologic coverage. The purpose of this manuscript is to provide a balanced view of the current state of the use of animal models and NAMs as approaches to development, safety, efficacy, and toxicity testing of drugs and chemicals. Animals do not provide all needed information nor do NAMs, but each can elucidate key pieces of the puzzle of human and animal biology and contribute to the goal of protecting human and animal health. SIGNIFICANCE STATEMENT: Data from traditional animal studies have predominantly been used to inform human health safety and efficacy. Although it is unlikely that all animal studies will be able to be replaced, with the continued advancement in new approach methods (NAMs), it is possible that sometime in the future, NAMs will likely be an important component by which the discovery, efficacy, and toxicity testing of drugs and chemicals is conducted and regulatory decisions are made.
Collapse
Affiliation(s)
- Barbara L F Kaplan
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Alan M Hoberman
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - William Slikker
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Mary Alice Smith
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Emanuela Corsini
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Thomas B Knudsen
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - M Sue Marty
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Sonya K Sobrian
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Suzanne C Fitzpatrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Marcia H Ratner
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Donna L Mendrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| |
Collapse
|
3
|
Cabrera-Najera LE, Chirino-Galindo G, Palomar-Morales M. Participation of lncRNAs in the development of diabetic complications: Systematic review and meta-analysis. I. Rat. Diabet Med 2024; 41:e15244. [PMID: 37846767 DOI: 10.1111/dme.15244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/08/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
AIMS We evaluated the involvement of lncRNAs in the development of pathologies associated with chronic hyperglycaemia in rat models in a model of type 1, type 2 and gestational diabetes. METHODS Reports were searched in Dialnet, Scielo, HINARI, Springer, ClinicalKey, OTseeker, PubMed and different grey literature databases with any restrictions. Bibliography databases will be searched from their inception to December 2022. RESULTS Thirty-seven studies met our criteria, and they had the following characteristics: original experimental studies on diabetes, the lncRNAs were extracted or measured from tissues of specific areas and the results were expressed in terms of standard measures by RT-PCR. In most studies, both primary and secondary outcomes were mentioned. On the other hand, we found a total of nine diabetic complications, being retinopathy, nephropathy and neuropathy the most representatives. Additionally, it was found that MALAT1, H19, NEAT1 and TUG1 are the most studied lncRNAs about these complications in rats. On the other hand, the lncRNAs with the highest rate of change were MSTRG.1662 (17.85; 13.78, 21.93), ENSRNOT00000093120_Aox3 (7.13; 5.95, 8.31) and NONRATG013497.2 (-5.55; -7.18, -3.93). CONCLUSIONS This review found a significant involvement of lncRNAs in the progression of pathologies associated with chronic hyperglycaemia in rat models, and further studies are needed to establish their potential as biomarkers and therapeutic targets for diabetes.
Collapse
Affiliation(s)
- Leonardo-Elias Cabrera-Najera
- Laboratorio de Metabolismo de la Diabetes Mellitus, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Gladys Chirino-Galindo
- Laboratorio de Metabolismo de la Diabetes Mellitus, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Martín Palomar-Morales
- Laboratorio de Metabolismo de la Diabetes Mellitus, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| |
Collapse
|
4
|
Brink CB, Lewis DI. The 12 Rs Framework as a Comprehensive, Unifying Construct for Principles Guiding Animal Research Ethics. Animals (Basel) 2023; 13:ani13071128. [PMID: 37048384 PMCID: PMC10093343 DOI: 10.3390/ani13071128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Animal research ethics and animal welfare in science have become progressively tightly regulated, and ethical integrity and scientific quality, as well as social responsiveness and responsibility have become key requirements for research to be approved, funded, published, and accepted. The multitude of factors to contemplate has in some instances not only become complex, requiring a team approach, but often perceived as confusing and overwhelming. To facilitate a process of simplistic yet comprehensive conceptualization, we developed the 12 Rs Framework to act as a mind map to guide scientists, oversight structures, and other stakeholders through the myriad of ethical considerations. It unfolds into three domains of twelve encompassing ethical principles, values, and other considerations, including the animal welfare, social values, and scientific integrity domains, whilst also recognizing the diversity of local context, legal requirements, values, and cultures around the globe. In the end, it can be seen as a unifying ethical framework to foster and promote animal research ethics.
Collapse
Affiliation(s)
- Christiaan B Brink
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa
| | - David I Lewis
- School of Biomedical Sciences & Biological Sciences Teaching Innovation Hub, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
5
|
MacVittie TJ. Where are the medical countermeasures against the ARS and DEARE? A current topic relative to an animal model research platform, radiation exposure context, the acute and delayed effects of acute exposure, and the FDA animal rule. Int J Radiat Biol 2023:1-15. [PMID: 36811500 DOI: 10.1080/09553002.2023.2181999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE A question echoed by the National Biodefense Science Board (NBSB) in 2010, remains a reasonable question in 2023; 'Where are the Countermeasures?'. A critical path for development of medical countermeasures (MCM) against acute, radiation-induced organ-specific injury within the acute radiation syndrome (ARS) and the delayed effects of acute radiation exposure (DEARE) requires the recognition of problems and solutions inherent in the path to FDA approval under the Animal Rule. Keep Rule number one in mind, It's not easy. CONSIDERATIONS The current topic herein is focused on defining the nonhuman primate model(s) for efficient MCM development relative to consideration of prompt and delayed exposure in the context of the nuclear scenario. The rhesus macaque is a predictive model for human exposure of partial-body irradiation with marginal bone marrow sparing that allows definition of the multiple organ injury in the acute radiation syndrome (ARS) and the delayed effects of acute radiation exposure (DEARE). The continued definition of natural history is required to delineate an associative or causal interaction within the concurrent multi-organ injury characteristic of the ARS and DEARE. A more efficient development of organ specific MCM for both pre-exposure and post-exposure prophylaxis to include acute radiation-induced combined injury requires closing critical gaps in knowledge and urgent support to rectify the national shortage of nonhuman primates. The rhesus macaque is a validated, predictive model of the human response to prompt and delayed radiation exposure, medical management and MCM treatment. A rational approach to further development of the cynomolgus macaque as a comparable model is urgently required for continued development of MCM for FDA approval. CONCLUSION It is imperative to examine the key variables relative to animal model development and validation, The pharmacokinetics, pharmacodynamics and exposure profiles, of candidate MCM relative to route, administration schedule and optimal efficacy define the fully effective dose. The conduct of adequate and well-controlled pivotal efficacy studies as well as safety and toxicity studies support approval under the FDA Animal Rule and label definition for human use.
Collapse
Affiliation(s)
- Thomas J MacVittie
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Schietgat L, Cuissart B, De Grave K, Efthymiadis K, Bureau R, Crémilleux B, Ramon J, Lepailleur A. Automated detection of toxicophores and prediction of mutagenicity using PMCSFG algorithm. Mol Inform 2023; 42:e2200232. [PMID: 36529710 DOI: 10.1002/minf.202200232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/23/2022]
Abstract
Maximum common substructures (MCS) have received a lot of attention in the chemoinformatics community. They are typically used as a similarity measure between molecules, showing high predictive performance when used in classification tasks, while being easily explainable substructures. In the present work, we applied the Pairwise Maximum Common Subgraph Feature Generation (PMCSFG) algorithm to automatically detect toxicophores (structural alerts) and to compute fingerprints based on MCS. We present a comparison between our MCS-based fingerprints and 12 well-known chemical fingerprints when used as features in machine learning models. We provide an experimental evaluation and discuss the usefulness of the different methods on mutagenicity data. The features generated by the MCS method have a state-of-the-art performance when predicting mutagenicity, while they are more interpretable than the traditional chemical fingerprints.
Collapse
Affiliation(s)
- Leander Schietgat
- Artificial Intelligence Lab, Vrije Universiteit Brussel, Brussel, Belgium.,Department of Computer Science, KU Leuven, Leuven, Belgium
| | - Bertrand Cuissart
- Groupe de Recherche en Informatique, Image, Automatique et Instrumentation de Caen, UNICAEN, ENSICAEN, CNRS - UMR GREYC, Normandie Univ., Caen, France
| | | | | | - Ronan Bureau
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, UNICAEN, CERMN, Normandie Univ., Caen, France
| | - Bruno Crémilleux
- Groupe de Recherche en Informatique, Image, Automatique et Instrumentation de Caen, UNICAEN, ENSICAEN, CNRS - UMR GREYC, Normandie Univ., Caen, France
| | - Jan Ramon
- INRIA Lille Nord Europe, Lille, France
| | - Alban Lepailleur
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, UNICAEN, CERMN, Normandie Univ., Caen, France
| |
Collapse
|
7
|
Begasse de Dhaem O, Wattiez AS, de Boer I, Pavitt S, Powers SW, Pradhan A, Gelfand AA, Nahman-Averbuch H. Bridging the gap between preclinical scientists, clinical researchers, and clinicians: From animal research to clinical practice. Headache 2023; 63:25-39. [PMID: 36633108 DOI: 10.1111/head.14441] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/02/2022] [Accepted: 08/26/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Collaborations amongst researchers and clinicians with complementary areas of expertise enhance knowledge for everyone and can lead to new discoveries. To facilitate these interactions, shared language and a general understanding of how colleagues in different subfields of headache and headache research approach their work are needed. METHODS This narrative review focuses on research methods applied in animal studies, human studies including clinical trials, and provides an overview of clinical practice. RESULTS For animal studies, we describe concepts needed to evaluate the quality and relevance of preclinical studies. For human research, fundamental concepts of neuroimaging, quantitative sensory testing, genetic and epidemiological research methods, and clinical research methodology that are commonly used in headache research are summarized. In addition, we provide an understanding of what guides headache clinicians, and summarize the practical approach to migraine management in adults and children. CONCLUSIONS It is hoped that this review facilitates further dialogue between clinicians and researchers that will help guide future research efforts and implementation of research findings into clinical practice.
Collapse
Affiliation(s)
| | - Anne-Sophie Wattiez
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, USA.,Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, Iowa, USA
| | - Irene de Boer
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Sara Pavitt
- Child & Adolescent Headache Program, University of California, San Francisco, California, USA
| | - Scott W Powers
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.,Center for Understanding Pediatric Pain, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Amynah Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amy A Gelfand
- Child & Adolescent Headache Program, University of California, San Francisco, California, USA
| | - Hadas Nahman-Averbuch
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Simulation Study of Radio Frequency Safety and the Optimal Size of a Single-Channel Surface Radio Frequency Coil for Mice at 9.4 T Magnetic Resonance Imaging. SENSORS 2022; 22:s22114274. [PMID: 35684895 PMCID: PMC9185248 DOI: 10.3390/s22114274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/22/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022]
Abstract
The optimized size of a single-channel surface radio frequency (RF) coil for mouse body images in a 9.4 T magnetic resonance imaging (MRI) system was determined via electromagnetic-field analysis of the signal depth according to the size of a single-channel coil. The single-channel surface RF coils used in electromagnetic field simulations were configured to operate in transmission/reception mode at a frequency of 9.4 T–400 MHz. Computational analysis using the finite-difference time-domain method was used to assess the single-channel surface RF coil by comparing single-channel surface RF coils of varying sizes in terms of |B1|-, |B1+|-, |B1−|- and |E|-field distribution. RF safety for the prevention of burn injuries to small animals was assessed using an analysis of the specific absorption rate. A single-channel surface RF coil with a 20 mm diameter provided optimal B1-field distribution and RF safety, thus confirming that single-channel surface RF coils with ≥25 mm diameter could not provide typical B1-field distribution. A single-channel surface RF coil with a 20 mm diameter for mouse body imaging at 9.4 T MRI was recommended to preserve the characteristics of single-channel surface RF coils, and ensured that RF signals were applied correctly to the target point within RF safety guidelines.
Collapse
|
9
|
Mancini C, Nannoni E. Relevance, Impartiality, Welfare and Consent: Principles of an Animal-Centered Research Ethics. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.800186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The principles of Replacement, Reduction and Refinement (3Rs) were developed to address the ethical dilemma that arises from the use of animals, without their consent, in procedures that may harm them but that are deemed necessary to achieve a greater good. While aiming to protect animals, the 3Rs are underpinned by a process-centered ethical perspective which regards them as instruments in a scientific apparatus. This paper explores the applicability of an animal-centered ethics to animal research, whereby animals would be regarded as autonomous subjects, legitimate stakeholders in and contributors to a research process, with their own interests and capable of consenting and dissenting to their involvement. This perspective derives from the ethical stance taken within the field of Animal-Computer Interaction (ACI), where researchers acknowledge that an animal-centered approach is essential to ensuring the best research outcomes. We propose the ethical principles of relevance, impartiality, welfare and consent, and a scoring system to help researchers and delegated authorities assess the extent to which a research procedure aligns with them. This could help researchers determine when being involved in research is indeed in an animal's best interests, when a procedure could be adjusted to increase its ethical standard or when the use of non-animal methods is more urgently advisable. We argue that the proposed principles should complement the 3Rs within an integrated ethical framework that recognizes animals' autonomy, interests and role, for a more nuanced ethical approach and for supporting the best possible research for the benefit animal partakers and wider society.
Collapse
|
10
|
Wang A, Madden LA, Paunov VN. Fabrication of Angiogenic Sprouting Coculture of Cell Clusteroids Using an Aqueous Two-Phase Pickering Emulsion System. ACS APPLIED BIO MATERIALS 2022; 5:1804-1816. [PMID: 35315278 DOI: 10.1021/acsabm.2c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumor cell spheroids and 3D cell culture have generated a lot of interest in the past decade due to their relative ease of production and biomedical research applications. To date, the frontier in tumor 3D models has been pushed to the level of personalized cancer treatment and customized tissue engineering applications. However, without vascularization, the central parts of these artificial constructs cannot survive without an adequate oxygen and nutrient supply. The formation of a necrotic core into in vitro 3D cell models still serves as the major obstacle in their wider practical application. Here, we propose a rapid formation protocol based on using a water-in-water (w/w) Pickering emulsion template to generate phenotypically endothelial/hepatic (ECV304/Hep-G2) coculture cell clusteroids with angiogenic capability. The w/w Pickering emulsion template was based on a dextran/poly(ethylene oxide) aqueous two-phase system stabilized by whey protein particles. The initial cell proportion in the coculture clusteroids can easily be manipulated for optimal performance. The cocultured pattern of the endothelial/hepatic cells could significantly promote the production of angiogenesis-related proteins. Our study confirmed that cocultured clusteroids can stimulate cell sprouting without the addition of vascular endothelial growth factor (VEGF) or other angiogenesis inducers at a 1:2 ratio of Hep-G2/ECV304. Angiogenesis gene production in the coculture clusteroids was enhanced with VEGF, urea, and insulin-like growth factor-binding protein along with angiogenesis-related marker CD34 levels, also indicating angiogenesis progress. Our aqueous two-phase Pickering emulsion templates provided a convenient approach to vascularize a target cell type in 3D cell coculture without additional stimulating factors, which could potentially apply to either cell lines or biopsy tissues, expanding the clusteroids downstream applications.
Collapse
Affiliation(s)
- Anheng Wang
- Department of Chemistry and Biochemistry, University of Hull, Hull HU67RX, United Kingdom
| | - Leigh A Madden
- Department of Biomedical Sciences, University of Hull, Hull HU67RX, United Kingdom
| | - Vesselin N Paunov
- Department of Chemistry, Nazarbayev University, 53 Kabanbay Batyr Avenue, Nursultan 010000, Kazakhstan
| |
Collapse
|
11
|
Ren J, Paxton NC, Hammond J, Saifzadeh S, Steck R, Lawrence FA, Woodruff MA. Novel resin tissue array system reduces sample preparation time, labour and reagent costs in bone tissue histology. Bone 2021; 153:116155. [PMID: 34411775 DOI: 10.1016/j.bone.2021.116155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022]
Abstract
Resin histology plays an essential role in the analysis of hard tissues, such as bone and teeth, as well as in the context of metallic implant analysis. However, the techniques of resin embedding, followed by ground sectioning, are very costly due to significantly increased reagent cost and labour time when compared to the conventional paraffin histology approach. In the present study, a novel resin array system was developed to increase the affordability of a project analysing rat femur tissues containing metallic or polymeric implants. The resin array system enabled the simultaneous embedding of the femur samples in groups of eight samples compared to the conventional resin method where samples are processed individually. The ground sections produced with the resin array system allowed uniform ROI selection, ground section thickness, staining consistency, and histological analysis with Goldner's trichrome stain, offering a substantial opportunity for reproducible immunohistochemistry which is unable to be achieved when processing samples embedded individually. The application of this novel resin array system significantly reduced resource usage when compared to doing the same analysis on individual samples. A reduction of approximately 40% was achieved for both total labour time and total reagent cost through the use of the array system compared with individual embedding. This novel resin array system has widespread applicability to many bone, hard tissue, and metallic implant studies, offering substantial conservation of research funds and increased accessibility to advanced techniques for commercial partners due to more cost-effective sample preparation and more accurate, reproducible data.
Collapse
Affiliation(s)
- Jiongyu Ren
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Naomi C Paxton
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Joshua Hammond
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Siamak Saifzadeh
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Roland Steck
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Felicity A Lawrence
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia
| | - Maria A Woodruff
- Queensland University of Technology (QUT), 2 George St, Brisbane, QLD 4000, Australia.
| |
Collapse
|
12
|
Species variations in tenocytes' response to inflammation require careful selection of animal models for tendon research. Sci Rep 2021; 11:12451. [PMID: 34127759 PMCID: PMC8203623 DOI: 10.1038/s41598-021-91914-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/24/2021] [Indexed: 01/23/2023] Open
Abstract
For research on tendon injury, many different animal models are utilized; however, the extent to which these species simulate the clinical condition and disease pathophysiology has not yet been critically evaluated. Considering the importance of inflammation in tendon disease, this study compared the cellular and molecular features of inflammation in tenocytes of humans and four common model species (mouse, rat, sheep, and horse). While mouse and rat tenocytes most closely equalled human tenocytes’ low proliferation capacity and the negligible effect of inflammation on proliferation, the wound closure speed of humans was best approximated by rats and horses. The overall gene expression of human tenocytes was most similar to mice under healthy, to horses under transient and to sheep under constant inflammatory conditions. Humans were best matched by mice and horses in their tendon marker and collagen expression, by horses in extracellular matrix remodelling genes, and by rats in inflammatory mediators. As no single animal model perfectly replicates the clinical condition and sufficiently emulates human tenocytes, fit-for-purpose selection of the model species for each specific research question and combination of data from multiple species will be essential to optimize translational predictive validity.
Collapse
|
13
|
The Role of GSK-3β in the Regulation of Protein Turnover, Myosin Phenotype, and Oxidative Capacity in Skeletal Muscle under Disuse Conditions. Int J Mol Sci 2021; 22:ijms22105081. [PMID: 34064895 PMCID: PMC8151958 DOI: 10.3390/ijms22105081] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscles, being one of the most abundant tissues in the body, are involved in many vital processes, such as locomotion, posture maintenance, respiration, glucose homeostasis, etc. Hence, the maintenance of skeletal muscle mass is crucial for overall health, prevention of various diseases, and contributes to an individual’s quality of life. Prolonged muscle inactivity/disuse (due to limb immobilization, mechanical ventilation, bedrest, spaceflight) represents one of the typical causes, leading to the loss of muscle mass and function. This disuse-induced muscle loss primarily results from repressed protein synthesis and increased proteolysis. Further, prolonged disuse results in slow-to-fast fiber-type transition, mitochondrial dysfunction and reduced oxidative capacity. Glycogen synthase kinase 3β (GSK-3β) is a key enzyme standing at the crossroads of various signaling pathways regulating a wide range of cellular processes. This review discusses various important roles of GSK-3β in the regulation of protein turnover, myosin phenotype, and oxidative capacity in skeletal muscles under disuse/unloading conditions and subsequent recovery. According to its vital functions, GSK-3β may represent a perspective therapeutic target in the treatment of muscle wasting induced by chronic disuse, aging, and a number of diseases.
Collapse
|
14
|
Razali K, Othman N, Mohd Nasir MH, Doolaanea AA, Kumar J, Ibrahim WN, Mohamed Ibrahim N, Mohamed WMY. The Promise of the Zebrafish Model for Parkinson's Disease: Today's Science and Tomorrow's Treatment. Front Genet 2021; 12:655550. [PMID: 33936174 PMCID: PMC8082503 DOI: 10.3389/fgene.2021.655550] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/23/2021] [Indexed: 11/29/2022] Open
Abstract
The second most prevalent neurodegenerative disorder in the elderly is Parkinson's disease (PD). Its etiology is unclear and there are no available disease-modifying medicines. Therefore, more evidence is required concerning its pathogenesis. The use of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is the basis of most animal models of PD. MPTP is metabolized by monoamine oxidase B (MAO B) to MPP + and induces the loss of dopaminergic neurons in the substantia nigra in mammals. Zebrafish have been commonly used in developmental biology as a model organism, but owing to its perfect mix of properties, it is now emerging as a model for human diseases. Zebrafish (Danio rerio) are cheap and easy to sustain, evolve rapidly, breed transparent embryos in large amounts, and are readily manipulated by different methods, particularly genetic ones. Furthermore, zebrafish are vertebrate species and mammalian findings obtained from zebrafish may be more applicable than those derived from genetic models of invertebrates such as Drosophila melanogaster and Caenorhabditis elegans. The resemblance cannot be taken for granted, however. The goal of the present review article is to highlight the promise of zebrafish as a PD animal model. As its aminergic structures, MPTP mode of action, and PINK1 roles mimic those of mammalians, zebrafish seems to be a viable model for studying PD. The roles of zebrafish MAO, however, vary from those of the two types of MAO present in mammals. The benefits unique to zebrafish, such as the ability to perform large-scale genetic or drug screens, should be exploited in future experiments utilizing zebrafish PD models.
Collapse
Affiliation(s)
- Khairiah Razali
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Noratikah Othman
- Department of Basic Medical Sciences, Kulliyyah of Nursing, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Mohd Hamzah Mohd Nasir
- Central Research and Animal Facility (CREAM), Kulliyyah of Science, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, UKM Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | | | - Wael M. Y. Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Menoufia, Egypt
| |
Collapse
|
15
|
Pai AC, Parekh KR, Engelhardt JF, Lynch TJ. Ferret respiratory disease models for the study of lung stem cells. LUNG STEM CELLS IN DEVELOPMENT, HEALTH AND DISEASE 2021:273-289. [DOI: 10.1183/2312508x.10010320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
16
|
Franco NH, Miranda SB, Kovács N, Nagy A, Thiện BQ, Reis F, Varga O. Assessing Scientific Soundness and Translational Value of Animal Studies on DPP4 Inhibitors for Treating Type 2 Diabetes Mellitus. BIOLOGY 2021; 10:155. [PMID: 33669354 PMCID: PMC7920304 DOI: 10.3390/biology10020155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022]
Abstract
Although there is a wide range of animal models of type 2 diabetes mellitus (T2DM) used in research; we have limited evidence on their translation value. This paper provides a) a comparison of preclinical animal and clinical results on the effect of five dipeptidyl peptidase-4 (DPP4) inhibitors by comparing the pharmaceutical caused glucose changes, and b) an evaluation of methodological and reporting standards in T2DM preclinical animal studies. DPP4 inhibitors play an important role in the clinical management of T2DM: if metformin alone is not sufficient enough to control the blood sugar levels, DPP4 inhibitors are often used as second-line therapy; additionally, DPP-4 inhibitors are also used in triple therapies with metformin and sodium-glucose co-transporter-2 (SGLT-2) inhibitors or with metformin and insulin. In our analysis of 124 preclinical studies and 47 clinical trials, (1) we found no evidence of species differences in glucose change response to DPP4 inhibitors, which may suggest that, for this drug class, studies in mice and rats may be equally predictive of how well a drug will work in humans; and (2) there is good reporting of group size, sex, age, euthanasia method and self-reported compliance with animal welfare regulations in animal studies but poor reporting of justification of group size, along with a strong bias towards the use of male animals and young animals. Instead of the common non-transparent model selection, we call for a reflective and evidenced-based assessment of predictive validity of the animal models currently available.
Collapse
Affiliation(s)
- Nuno Henrique Franco
- Laboratory Animal Science Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (N.H.F.); (S.B.M.)
- Instituto de Investigação e Inovação da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Sonia Batista Miranda
- Laboratory Animal Science Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (N.H.F.); (S.B.M.)
- Instituto de Investigação e Inovação da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Nóra Kovács
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Kassai út 26, 4028 Debrecen, Hungary;
| | - Attila Nagy
- Faculty of Public Health, University of Debrecen, Kassai út 26, 4028 Debrecen, Hungary;
| | - Bùi Quốc Thiện
- Faculty of Medicine, University of Debrecen, Egyetem Square 1, 4032 Debrecen, Hungary;
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Orsolya Varga
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Kassai út 26, 4028 Debrecen, Hungary;
- Office for Research Groups Attached to Universities and Other Institutions, Hungarian Academy of Sciences, 1051 Budapest, Hungary
| |
Collapse
|
17
|
Wang MWH, Goodman JM, Allen TEH. Machine Learning in Predictive Toxicology: Recent Applications and Future Directions for Classification Models. Chem Res Toxicol 2020; 34:217-239. [PMID: 33356168 DOI: 10.1021/acs.chemrestox.0c00316] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent times, machine learning has become increasingly prominent in predictive toxicology as it has shifted from in vivo studies toward in silico studies. Currently, in vitro methods together with other computational methods such as quantitative structure-activity relationship modeling and absorption, distribution, metabolism, and excretion calculations are being used. An overview of machine learning and its applications in predictive toxicology is presented here, including support vector machines (SVMs), random forest (RF) and decision trees (DTs), neural networks, regression models, naïve Bayes, k-nearest neighbors, and ensemble learning. The recent successes of these machine learning methods in predictive toxicology are summarized, and a comparison of some models used in predictive toxicology is presented. In predictive toxicology, SVMs, RF, and DTs are the dominant machine learning methods due to the characteristics of the data available. Lastly, this review describes the current challenges facing the use of machine learning in predictive toxicology and offers insights into the possible areas of improvement in the field.
Collapse
Affiliation(s)
- Marcus W H Wang
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Jonathan M Goodman
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Timothy E H Allen
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,MRC Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, Leicester LE1 7HB, United Kingdom
| |
Collapse
|
18
|
Determining effects of adolescent stress exposure on risk for posttraumatic stress disorder in adulthood. Curr Opin Behav Sci 2020. [DOI: 10.1016/j.cobeha.2020.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Rikhi R, Samra G, Arustamyan M, Patel J, Zhou L, Bungo B, Moudgil R. Radiation induced cardiovascular disease: An odyssey of bedside-bench-bedside approach. LIFE SCIENCES IN SPACE RESEARCH 2020; 27:49-55. [PMID: 34756229 DOI: 10.1016/j.lssr.2020.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 06/13/2023]
Abstract
The journey to Mars will be an ambitious, yet arduous task as it will entail culmination of all the information we have gathered over many decades. While the mission is of utmost importance, preservation of astronaut's well-being is paramount also. To that end, mitigation of radiation risk especially afflicting cardiovascular disease (CVD) is of great interest and challenge. Current data from astronauts on low earth orbit and Apollo missions provides insight on the risk of CVD from radiation exposure. However, data is limited given the small cohort size of astronauts who embarked on just nine prolonged missions. Therefore, a cerebral approach to understanding and mitigating risks are essential. This paper discusses the need for a predictive preclinical model to help understand and mitigate the effects of radiation on astronauts. We will discuss strengths and limitations of preclinical models and the methods of validating and constructing a model to predict human clinical outcomes. Our bedside-bench-bedside approach focuses on adapting the preclinical model through common investigative tools used between humans and animals. The result will be an optimization of preclinical model to a point of being a surrogate clinical model capable of predicting CVD outcomes in astronauts exposed to radiation.
Collapse
Affiliation(s)
- Rishi Rikhi
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Gursharan Samra
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Michael Arustamyan
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Jay Patel
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Leon Zhou
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Brandon Bungo
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Rohit Moudgil
- Section of Clinical Cardiology, Department of Cardiovascular Medicine Heart and Vascular Institute Cleveland Clinic Foundation 9500 Euclid Ave, Cleveland, OH 44195, USA.
| |
Collapse
|
20
|
Ferreira GS, Veening-Griffioen DH, Boon WPC, Moors EHM, van Meer PJK. Levelling the Translational Gap for Animal to Human Efficacy Data. Animals (Basel) 2020; 10:E1199. [PMID: 32679706 PMCID: PMC7401509 DOI: 10.3390/ani10071199] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Reports of a reproducibility crisis combined with a high attrition rate in the pharmaceutical industry have put animal research increasingly under scrutiny in the past decade. Many researchers and the general public now question whether there is still a justification for conducting animal studies. While criticism of the current modus operandi in preclinical research is certainly warranted, the data on which these discussions are based are often unreliable. Several initiatives to address the internal validity and reporting quality of animal studies (e.g., Animals in Research: Reporting In Vivo Experiments (ARRIVE) and Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE) guidelines) have been introduced but seldom implemented. As for external validity, progress has been virtually absent. Nonetheless, the selection of optimal animal models of disease may prevent the conducting of clinical trials, based on unreliable preclinical data. Here, we discuss three contributions to tackle the evaluation of the predictive value of animal models of disease themselves. First, we developed the Framework to Identify Models of Disease (FIMD), the first step to standardise the assessment, validation and comparison of disease models. FIMD allows the identification of which aspects of the human disease are replicated in the animals, facilitating the selection of disease models more likely to predict human response. Second, we show an example of how systematic reviews and meta-analyses can provide another strategy to discriminate between disease models quantitatively. Third, we explore whether external validity is a factor in animal model selection in the Investigator's Brochure (IB), and we use the IB-derisk tool to integrate preclinical pharmacokinetic and pharmacodynamic data in early clinical development. Through these contributions, we show how we can address external validity to evaluate the translatability and scientific value of animal models in drug development. However, while these methods have potential, it is the extent of their adoption by the scientific community that will define their impact. By promoting and adopting high quality study design and reporting, as well as a thorough assessment of the translatability of drug efficacy of animal models of disease, we will have robust data to challenge and improve the current animal research paradigm.
Collapse
Affiliation(s)
- Guilherme S. Ferreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands; (D.H.V.-G.); (P.J.K.v.M.)
| | - Désirée H. Veening-Griffioen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands; (D.H.V.-G.); (P.J.K.v.M.)
| | - Wouter P. C. Boon
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, 3512 JE Utrecht, The Netherlands; (W.P.C.B.); (E.H.M.M.)
| | - Ellen H. M. Moors
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, 3512 JE Utrecht, The Netherlands; (W.P.C.B.); (E.H.M.M.)
| | - Peter J. K. van Meer
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands; (D.H.V.-G.); (P.J.K.v.M.)
- Medicines Evaluation Board, 3531 AH Utrecht, The Netherlands
| |
Collapse
|
21
|
Golenser J, Salaymeh N, Higazi AA, Alyan M, Daif M, Dzikowski R, Domb AJ. Treatment of Experimental Cerebral Malaria by Slow Release of Artemisone From Injectable Pasty Formulation. Front Pharmacol 2020; 11:846. [PMID: 32595499 PMCID: PMC7303303 DOI: 10.3389/fphar.2020.00846] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/22/2020] [Indexed: 12/26/2022] Open
Abstract
Malaria caused by Plasmodium falciparum causes numerous cases of morbidity with about 400,000 deaths yearly owing, mainly, to inflammation leading to cerebral malaria (CM). CM conventionally is treated by repetitive administration of anti-plasmodial drugs and supportive non-specific drugs, for about a week. A mouse model of CM caused by Plasmodium berghei ANKA, in which brain and systemic clinical pathologies occur followed by sudden death within about a week, was used to study the effect of artemisone, a relatively new artemisinin, within an injectable pasty polymer formulated for its controlled release. The parasites were exposed to the drug over several days at a non-toxic concentrations for the mice but high enough to affect the parasites. Artemisone was also tested in cultures of bacteria, cancer cells and P. falciparum to evaluate the specificity and suitability of these cells for examining the release of artemisone from its carrier. Cultures of P. falciparum were the most suitable. Artemisone released from subcutaneous injected poly(sebacic acid-ricinoleic acid) (PSARA) pasty polymer, reduced parasitemias in infected mice, prolonged survival and prevented death in most of the infected mice. Successful prophylactic treatment before infection proved that there was a slow release of the drug for about a week, which contrasts with the three hour half-life that occurs after injection of just the drug. Treatment with artemisone within the polymer, even at a late stage of the disease, helped to prevent or, at least, delay accompanying severe symptoms. In some cases, treatment prevented death of CM and the mice died later of anemia. Postponing the severe clinical symptoms is also beneficial in cases of human malaria, giving more time for an appropriate diagnosis and treatment before severe symptoms appear. The method presented here may also be useful for combination therapy of anti-plasmodial and immunomodulatory drugs.
Collapse
Affiliation(s)
- Jacob Golenser
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | - Nadeen Salaymeh
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | | | - Mohammed Alyan
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| | - Mahran Daif
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| | - Ron Dzikowski
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | - Abraham J. Domb
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| |
Collapse
|
22
|
A critical inquiry into marble-burying as a preclinical screening paradigm of relevance for anxiety and obsessive-compulsive disorder: Mapping the way forward. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2020; 19:1-39. [PMID: 30361863 DOI: 10.3758/s13415-018-00653-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rodent marble-burying behavior in the marble-burying test (MBT) is employed as a model or measure to study anxiety- and compulsive-like behaviors or anxiolytic and anticompulsive drug action. However, the test responds variably to a range of pharmacological interventions, and little consensus exists regarding specific methodologies for its execution. Regardless, the test is widely applied to investigate the effects of pharmacological, genetic, and behavioral manipulations on purported behaviors related to the said neuropsychiatric constructs. Therefore, in the present review we attempt to expound the collective translational significance of the MBT. We do this by (1) reviewing burying behavior as a natural behavioral phenotype, (2) highlighting key aspects of anxiety and obsessive-compulsive disorder from a translational perspective, (3) reviewing the history and proof of concept of the MBT, (4) critically appraising potential methodological confounds in execution of the MBT, and (5) dissecting responses of the MBT to various pharmacological interventions. We conclude by underlining that the collective translational value of the MBT will be strengthened by contextually valid experimental designs and objective reporting of data.
Collapse
|
23
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
24
|
Miccoli B, Braeken D, Li YCE. Brain-on-a-chip Devices for Drug Screening and Disease Modeling Applications. Curr Pharm Des 2019; 24:5419-5436. [PMID: 30806304 DOI: 10.2174/1381612825666190220161254] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are related to the progressive functional loss of the brain, often connected to emotional and physical disability and, ultimately, to death. These disorders, strongly connected to the aging process, are becoming increasingly more relevant due to the increase of life expectancy. Current pharmaceutical treatments poorly tackle these diseases, mainly acting only on their symptomology. One of the main reasons of this is the current drug development process, which is not only expensive and time-consuming but, also, still strongly relies on animal models at the preclinical stage. Organ-on-a-chip platforms have the potential to strongly impact and improve the drug screening process by recreating in vitro the functionality of human organs. Patient-derived neurons from different regions of the brain can be directly grown and differentiated on a brain-on-a-chip device where the disease development, progression and pharmacological treatments can be studied and monitored in real time. The model reliability is strongly improved by using human-derived cells, more relevant than animal models for pharmacological screening and disease monitoring. The selected cells will be then capable of proliferating and organizing themselves in the in vivo environment thanks to the device architecture, materials selection and bio-chemical functionalization. In this review, we start by presenting the fundamental strategies adopted for brain-on-a-chip devices fabrication including e.g., photolithography, micromachining and 3D printing technology. Then, we discuss the state-of-theart of brain-on-a-chip platforms including their role in the study of the functional architecture of the brain e.g., blood-brain barrier, or of the most diffuse neurodegenerative diseases like Alzheimer's and Parkinson's. At last, the current limitations and future perspectives of this approach for the development of new drugs and neurodegenerative diseases modeling will be discussed.
Collapse
Affiliation(s)
- Beatrice Miccoli
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium.,Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dries Braeken
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium
| | - Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung City, Taiwan
| |
Collapse
|
25
|
S. Ferreira G, Veening-Griffioen DH, Boon WPC, Moors EHM, Gispen-de Wied CC, Schellekens H, van Meer PJK. A standardised framework to identify optimal animal models for efficacy assessment in drug development. PLoS One 2019; 14:e0218014. [PMID: 31194784 PMCID: PMC6563989 DOI: 10.1371/journal.pone.0218014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/23/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Poor translation of efficacy data derived from animal models can lead to clinical trials unlikely to benefit patients-or even put them at risk-and is a potential contributor to costly and unnecessary attrition in drug development. OBJECTIVES To develop a tool to assess, validate and compare the clinical translatability of animal models used for the preliminary assessment of efficacy. DESIGN AND RESULTS We performed a scoping review to identify the key aspects used to validate animal models. Eight domains (Epidemiology, Symptomatology and Natural History-SNH, Genetic, Biochemistry, Aetiology, Histology, Pharmacology and Endpoints) were identified. We drafted questions to evaluate the different facets of human disease simulation. We designed the Framework to Identify Models of Disease (FIMD) to include standardised instructions, a weighting and scoring system to compare models as well as factors to help interpret model similarity and evidence uncertainty. We also added a reporting quality and risk of bias assessment of drug intervention studies in the Pharmacological Validation domain. A web-based survey was conducted with experts from different stakeholders to gather input on the framework. We conducted a pilot study of the validation in two models for Type 2 Diabetes (T2D)-the ZDF rat and db/db mouse. Finally, we present a full validation and comparison of two animal models for Duchenne Muscular Dystrophy (DMD): the mdx mouse and GRMD dog. We show that there are significant differences between the mdx mouse and the GRMD dog, the latter mimicking the human epidemiological, SNH, and histological aspects to a greater extent than the mouse despite the overall lack of published data. CONCLUSIONS FIMD facilitates drug development by serving as the basis to select the most relevant model that can provide meaningful data and is more likely to generate translatable results to progress drug candidates to the clinic.
Collapse
Affiliation(s)
- Guilherme S. Ferreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Désirée H. Veening-Griffioen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wouter P. C. Boon
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | - Ellen H. M. Moors
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | | | - Huub Schellekens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Peter J. K. van Meer
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Medicines Evaluation Board, Utrecht, The Netherlands
| |
Collapse
|
26
|
Kuna L, Bozic I, Kizivat T, Bojanic K, Mrso M, Kralj E, Smolic R, Wu GY, Smolic M. Models of Drug Induced Liver Injury (DILI) - Current Issues and Future Perspectives. Curr Drug Metab 2018; 19:830-838. [PMID: 29788883 PMCID: PMC6174638 DOI: 10.2174/1389200219666180523095355] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Background: Drug-induced Liver Injury (DILI) is an important cause of acute liver failure cases in the United States, and remains a common cause of withdrawal of drugs in both preclinical and clinical phases. Methods: A structured search of bibliographic databases – Web of Science Core Collection, Scopus and Medline for peer-reviewed articles on models of DILI was performed. The reference lists of relevant studies was prepared and a citation search for the included studies was carried out. In addition, the characteristics of screened studies were described. Results: One hundred and six articles about the existing knowledge of appropriate models to study DILI in vitro and in vivo with special focus on hepatic cell models, variations of 3D co-cultures, animal models, databases and predictive modeling and translational biomarkers developed to understand the mechanisms and pathophysiology of DILI are described. Conclusion: Besides descriptions of current applications of existing modeling systems, associated advantages and limitations of each modeling system and future directions for research development are discussed as well.
Collapse
Affiliation(s)
- Lucija Kuna
- Department of Chemistry and Biochemistry, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Ivana Bozic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Tomislav Kizivat
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Kristina Bojanic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Margareta Mrso
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - Edgar Kralj
- Inspecto, LLC, Martina Divalta 193, 31000 Osijek, Croatia
| | - Robert Smolic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia
| | - George Y Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, United States
| | - Martina Smolic
- Department of Pharmacology, Faculty of Medicine, J. J. Strossmayer University of Osijek, J. Huttlera 4, 31000 Osijek, Croatia.,Department of Pharmacology, Faculty Of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| |
Collapse
|
27
|
McCutcheon V, Park E, Liu E, Sobhebidari P, Tavakkoli J, Wen XY, Baker AJ. A Novel Model of Traumatic Brain Injury in Adult Zebrafish Demonstrates Response to Injury and Treatment Comparable with Mammalian Models. J Neurotrauma 2016; 34:1382-1393. [PMID: 27650063 DOI: 10.1089/neu.2016.4497] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and morbidity in industrialized countries with considerable associated health care costs. The cost and time associated with pre-clinical development of TBI therapeutics is lengthy and expensive with a poor track record of successful translation to the clinic. The zebrafish is an emerging model organism in research with unique technical and genomic strengths in the study of disease and development. Its high degree of genetic homology and cell signaling pathways relative to mammalian species and amenability to high and medium throughput assays has potential to accelerate the rate of therapeutic drug identification. Accordingly, we developed a novel closed-head model of TBI in adult zebrafish using a targeted, pulsed, high-intensity focused ultrasound (pHIFU) to induce mechanical injury of the brain. Western blot results indicated altered microtubule and neurofilament expression as well as increased expression of cleaved caspase-3 and beta APP (β-APP; p < 0.05). We used automated behavioral tracking software to evaluate locomotor deficits 24 and 48 h post-injury. Significant behavioral impairment included decreased swim distance and velocity (p < 0.05), as well as heightened anxiety and altered group social dynamics. Responses to injury were pHIFU dose-dependent and modifiable with MK-801, MDL-28170, or temperature modulation. Together, results indicate that the zebrafish exhibits responses to injury and intervention similar to mammalian TBI pathophysiology and suggest the potential for use to rapidly evaluate therapeutic compounds with high efficiency.
Collapse
Affiliation(s)
| | - Eugene Park
- 2 Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario, Canada
| | - Elaine Liu
- 2 Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario, Canada
| | - Pooya Sobhebidari
- 3 Department of Physics, Ryerson University , Toronto, Ontario, Canada
| | - Jahan Tavakkoli
- 2 Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario, Canada .,3 Department of Physics, Ryerson University , Toronto, Ontario, Canada
| | - Xiao-Yan Wen
- 2 Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario, Canada .,4 Departments of Medicine and Physiology, University of Toronto , Ontario, Canada
| | - Andrew J Baker
- 1 Institute of Medical Sciences, University of Toronto , Ontario, Canada .,2 Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario, Canada .,5 Departments of Anesthesia and Surgery, University of Toronto , Ontario, Canada
| |
Collapse
|
28
|
Raess M, de Castro AA, Gailus-Durner V, Fessele S, Hrabě de Angelis M. INFRAFRONTIER: a European resource for studying the functional basis of human disease. Mamm Genome 2016; 27:445-50. [PMID: 27262858 PMCID: PMC4935733 DOI: 10.1007/s00335-016-9642-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/14/2016] [Indexed: 12/20/2022]
Abstract
Ageing research and more generally the study of the functional basis of human diseases profit enormously from the large-scale approaches and resources in mouse functional genomics: systematic targeted mutation of the mouse genome, systemic phenotyping in mouse clinics, and the archiving and distribution of the mouse resources in public repositories. INFRAFRONTIER, the European research infrastructure for the development, systemic phenotyping, archiving and distribution of mammalian models, offers access to sustainable mouse resources for biomedical research. INFRAFRONTIER promotes the global sharing of high-quality resources and data and thus contributes to data reproducibility and animal welfare. INFRAFRONTIER puts great effort into international standardisation and quality control and into technology development to improve and expand experimental protocols, reduce the use of animals in research and increase the reproducibility of results. In concert with the research community and the International Mouse Phenotyping Consortium (IMPC), INFRAFRONTIER is currently developing new pilot platforms and services for the research on ageing and age-related diseases.
Collapse
Affiliation(s)
| | | | - Valérie Gailus-Durner
- Institute of Experimental Genetics & German Mouse Clinic, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | | | - Martin Hrabě de Angelis
- INFRAFRONTIER GmbH, 85764, Neuherberg, Germany.
- Institute of Experimental Genetics & German Mouse Clinic, Helmholtz Zentrum München, 85764, Neuherberg, Germany.
| |
Collapse
|
29
|
Fifel K, Piggins H, Deboer T. Modeling sleep alterations in Parkinson's disease: How close are we to valid translational animal models? Sleep Med Rev 2016; 25:95-111. [DOI: 10.1016/j.smrv.2015.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 10/23/2022]
|
30
|
Rafiq QA, Ortega I, Jenkins SI, Wilson SL, Patel AK, Barnes AL, Adams CF, Delcassian D, Smith D. The early career researcher's toolkit: translating tissue engineering, regenerative medicine and cell therapy products. Regen Med 2015; 10:989-1003. [PMID: 26628407 DOI: 10.2217/rme.15.56] [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: 11/21/2022] Open
Abstract
Although the importance of translation for the development of tissue engineering, regenerative medicine and cell-based therapies is widely recognized, the process of translation is less well understood. This is particularly the case among some early career researchers who may not appreciate the intricacies of translational research or make decisions early in development which later hinders effective translation. Based on our own research and experiences as early career researchers involved in tissue engineering and regenerative medicine translation, we discuss common pitfalls associated with translational research, providing practical solutions and important considerations which will aid process and product development. Suggestions range from effective project management, consideration of key manufacturing, clinical and regulatory matters and means of exploiting research for successful commercialization.
Collapse
Affiliation(s)
- Qasim A Rafiq
- Centre for Biological Engineering, Wolfson School of Mechanical & Manufacturing Engineering, Loughborough University, Leicestershire, LE11 3TU, UK.,Aston Medical Research Institute, School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Ilida Ortega
- Bioengineering & Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, S10 2TA, UK
| | - Stuart I Jenkins
- Institute for Science & Technology in Medicine, Keele University, Staffordshire, ST5 5BG, UK
| | - Samantha L Wilson
- Academic Ophthalmology, Division of Clincial Neuroscience, Queen's Medical Centre Campus, University of Nottingham, NG7 2UH, UK
| | - Asha K Patel
- Wolfson Centre for Stem Cells, Tissue Engineering & Modeling, University of Nottingham, Nottingham, NG7 2RD, UK.,David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Christopher F Adams
- Institute for Science & Technology in Medicine, Keele University, Staffordshire, ST5 5BG, UK
| | - Derfogail Delcassian
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK.,Wolfson Centre for Stem Cells, Centre for Biological Sciences, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - David Smith
- Centre for Biological Engineering, Wolfson School of Mechanical & Manufacturing Engineering, Loughborough University, Leicestershire, LE11 3TU, UK.,PCT, a Caladrius company, 4 Pearl Court, Suite C, Allendale, NJ 07401, USA
| |
Collapse
|
31
|
Green SB. Can animal data translate to innovations necessary for a new era of patient-centred and individualised healthcare? Bias in preclinical animal research. BMC Med Ethics 2015. [PMID: 26215508 PMCID: PMC4517563 DOI: 10.1186/s12910-015-0043-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background The public and healthcare workers have a high expectation of animal research which they perceive as necessary to predict the safety and efficacy of drugs before testing in clinical trials. However, the expectation is not always realised and there is evidence that the research often fails to stand up to scientific scrutiny and its 'predictive value' is either weak or absent. Discussion Problems with the use of animals as models of humans arise from a variety of biases and systemic failures including: 1) bias and poor practice in research methodology and data analysis; 2) lack of transparency in scientific assessment and regulation of the research; 3) long-term denial of weaknesses in cross-species translation; 4) profit-driven motives overriding patient interests; 5) lack of accountability of expenditure on animal research; 6) reductionist-materialism in science which tends to dictate scientific inquiry and control the direction of funding in biomedical research. Summary Bias in animal research needs to be addressed before medical research and healthcare decision-making can be more evidence-based. Research funding may be misdirected on studying 'disease mechanisms' in animals that cannot be replicated outside tightly controlled laboratory conditions, and without sufficient critical evaluation animal research may divert attention away from avenues of research that hold promise for human health. The potential for harm to patients and trial volunteers from reliance on biased animal data1 requires measures to improve its conduct, regulation and analysis. This article draws attention to a few of the many forms of bias in animal research that have come to light in the last decade and offers a strategy incorporating ten recommendations stated at the end of each section on bias. The proposals need development through open debate and subsequent rigorous implementation so that reviewers may determine the value of animal research to human health. The 10Rs + are protected by a Creative Commons Attribution 3.0 Unported License and therefore may be 'shared, remixed or built on, even commercially, so long as attributed by giving appropriate credit with a link to the license, and indicate if changes were made.’ Electronic supplementary material The online version of this article (doi:10.1186/s12910-015-0043-7) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Zuluaga AF, Salazar BE, Agudelo M, Rodriguez CA, Vesga O. A strain-independent method to induce progressive and lethal pneumococcal pneumonia in neutropenic mice. J Biomed Sci 2015; 22:24. [PMID: 25890037 PMCID: PMC4474571 DOI: 10.1186/s12929-015-0124-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/26/2015] [Indexed: 11/10/2022] Open
Abstract
Background Experimental models of pneumonia with penicillin non-susceptible Streptococcus pneumoniae (PNSSP) are hard to reproduce because the majority of strains with clinical relevance (like serotypes 6B, 9 V and 19 F) have low murine virulence. By optimization of culture and inoculum conditions of PNSSP (using porcine mucin), our aim was to develop a suitable, reliable and reproducible pneumonia mouse model for anti-infective pharmacology research. Results Seven PNSSP strains, including serotypes 6B, 9 V, 14 and 19 F were included. Strain INS-E611 displayed the highest murine virulence and was chosen to validate the lung model. Nose-instilled pneumococci grew between 2.1 and 2.5 log10 CFU/g of lung in 24 hours when an optimized culture of bacterial cells was used, but animals were all alive and recovered of infection after 36 h. In contrast, inoculum supplementation with mucin led to 100% mortality related to a successful lung infection confirmed by histopathology. These findings were reproduced with all seven PNSSP strains in neutropenic mice. Immunocompetent animals cleared all strains spontaneously. Conclusions This pneumonia model produces a progressive and uniformly fatal lung infection with diverse serotypes of PNSSP independently of their intrinsic murine virulence.
Collapse
Affiliation(s)
- Andres F Zuluaga
- GRIPE [Grupo Investigador de Problemas en Enfermedades infecciosas], Medellín, Colombia. .,Department of Pharmacology and Toxicology, Medellín, Colombia.
| | | | - Maria Agudelo
- GRIPE [Grupo Investigador de Problemas en Enfermedades infecciosas], Medellín, Colombia. .,Department of Pharmacology and Toxicology, Medellín, Colombia. .,Infectious Diseases Unit, Hospital Universitario San Vicente Fundación, Medellín, Colombia.
| | - Carlos A Rodriguez
- GRIPE [Grupo Investigador de Problemas en Enfermedades infecciosas], Medellín, Colombia. .,Department of Pharmacology and Toxicology, Medellín, Colombia.
| | - Omar Vesga
- GRIPE [Grupo Investigador de Problemas en Enfermedades infecciosas], Medellín, Colombia. .,Department of Pharmacology and Toxicology, Medellín, Colombia. .,Department of Internal Medicine, Universidad de Antioquia, Calle 70 No. 52-21, Medellín, Colombia. .,Infectious Diseases Unit, Hospital Universitario San Vicente Fundación, Medellín, Colombia.
| |
Collapse
|
33
|
Ibrahim MM, Bond J, Bergeron A, Miller KJ, Ehanire T, Quiles C, Lorden ER, Medina MA, Fisher M, Klitzman B, Selim MA, Leong KW, Levinson H. A novel immune competent murine hypertrophic scar contracture model: a tool to elucidate disease mechanism and develop new therapies. Wound Repair Regen 2015; 22:755-64. [PMID: 25327261 PMCID: PMC4304906 DOI: 10.1111/wrr.12238] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 09/04/2014] [Indexed: 01/06/2023]
Abstract
Hypertrophic scar (HSc) contraction following burn injury causes contractures. Contractures are painful and disfiguring. Current therapies are marginally effective. To study pathogenesis and develop new therapies, a murine model is needed. We have created a validated immune-competent murine HSc model. A third-degree burn was created on dorsum of C57BL/6 mice. Three days postburn, tissue was excised and grafted with ear skin. Graft contraction was analyzed and tissue harvested on different time points. Outcomes were compared with human condition to validate the model. To confirm graft survival, green fluorescent protein (GFP) mice were used, and histologic analysis was performed to differentiate between ear and back skin. Role of panniculus carnosus in contraction was analyzed. Cellularity was assessed with 4′,6-diamidino-2-phenylindole. Collagen maturation was assessed with Picro-sirius red. Mast cells were stained with Toluidine blue. Macrophages were detected with F4/80 immune. Vascularity was assessed with CD31 immune. RNA for contractile proteins was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Elastic moduli of skin and scar tissue were analyzed using a microstrain analyzer. Grafts contracted to ∼45% of their original size by day 14 and maintained their size. Grafting of GFP mouse skin onto wild-type mice, and analysis of dermal thickness and hair follicle density, confirmed graft survival. Interestingly, hair follicles disappeared after grafting and regenerated in ear skin configuration by day 30. Radiological analysis revealed that panniculus carnosus doesn't contribute to contraction. Microscopic analyses showed that grafts show increase in cellularity. Granulation tissue formed after day 3. Collagen analysis revealed increases in collagen maturation over time. CD31 stain revealed increased vascularity. Macrophages and mast cells were increased. qRT-PCR showed up-regulation of transforming growth factor beta, alpha smooth muscle actin, and rho-associated protein kinase 2 in HSc. Tensile testing revealed that human skin and scar tissues are tougher than mouse skin and scar tissues.
Collapse
Affiliation(s)
- Mohamed Magdy Ibrahim
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
An optimized mouse thigh infection model for enterococci and its impact on antimicrobial pharmacodynamics. Antimicrob Agents Chemother 2014; 59:233-8. [PMID: 25348523 DOI: 10.1128/aac.02352-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Negligible in vivo growth of enterococci and high-level dispersion of data have led to inaccurate estimations of antibiotic pharmacodynamics (PD). Here we improved an in vivo model apt for PD studies by optimizing the in vitro culture conditions for enterococci. The PD of vancomycin (VAN), ampicillin-sulbactam (SAM), and piperacillin-tazobactam (TZP) against enterococci were determined in vivo, comparing the following different conditions of inoculum preparation: aerobiosis, aerobiosis plus mucin, and anaerobiosis plus mucin. Drug exposure was expressed as the ratio of the area under the concentration-time curve for the free, unbound fraction of the drug to the MIC (fAUC/MIC) (VAN) or the time in a 24-h period that the drug concentration for the free, unbound fraction exceeded the MIC under steady-state pharmacokinetic conditions (fT(>MIC)) (SAM and TZP) and linked to the change in log10 CFU/thigh. Only anaerobiosis plus mucin enhanced the in vivo growth, yielding significant PD parameters with all antibiotics. In conclusion, robust in vivo growth of enterococci was crucial for better determining the PD of tested antibacterial agents, and this was achieved by optimizing the procedure for preparing the inoculum.
Collapse
|
35
|
Zuluaga AF, Rodriguez CA, Agudelo M, Vesga O. About the validation of animal models to study the pharmacodynamics of generic antimicrobials. Clin Infect Dis 2014; 59:459-61. [PMID: 24785234 DOI: 10.1093/cid/ciu306] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Andres F Zuluaga
- Grupo Investigador de Problemas en Enfermedades Infecciosas (GRIPE) Department of Pharmacology and Toxicology
| | - Carlos A Rodriguez
- Grupo Investigador de Problemas en Enfermedades Infecciosas (GRIPE) Department of Pharmacology and Toxicology
| | - Maria Agudelo
- Grupo Investigador de Problemas en Enfermedades Infecciosas (GRIPE) Department of Pharmacology and Toxicology
| | - Omar Vesga
- Grupo Investigador de Problemas en Enfermedades Infecciosas (GRIPE) Department of Pharmacology and Toxicology Department of Internal Medicine, School of Medicine, Universidad de Antioquia Department of Infectious Diseases Unit, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| |
Collapse
|
36
|
Interspecies differences with in vitro and in vivo models of vascular tissue engineering. Biomaterials 2013; 34:9842-52. [DOI: 10.1016/j.biomaterials.2013.07.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/26/2013] [Indexed: 11/30/2022]
|
37
|
Moroni M, Ngudiankama BF, Christensen C, Olsen CH, Owens R, Lombardini ED, Holt RK, Whitnall MH. The Gottingen minipig is a model of the hematopoietic acute radiation syndrome: G-colony stimulating factor stimulates hematopoiesis and enhances survival from lethal total-body γ-irradiation. Int J Radiat Oncol Biol Phys 2013; 86:986-92. [PMID: 23845847 DOI: 10.1016/j.ijrobp.2013.04.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE We are characterizing the Gottingen minipig as an additional large animal model for advanced drug testing for the acute radiation syndrome (ARS) to enhance the discovery and development of novel radiation countermeasures. Among the advantages provided by this model, the similarities to human hematologic parameters and dynamics of cell loss/recovery after irradiation provide a convenient means to compare the efficacy of drugs known to affect bone marrow cellularity and hematopoiesis. METHODS AND MATERIALS Male Gottingen minipigs, 4 to 5 months old and weighing 9 to 11 kg, were used for this study. We tested the standard off-label treatment for ARS, rhG-CSF (Neupogen, 10 μg/kg/day for 17 days), at the estimated LD70/30 total-body γ-irradiation (TBI) radiation dose for the hematopoietic syndrome, starting 24 hours after irradiation. RESULTS The results indicated that granulocyte colony stimulating factor (G-CSF) enhanced survival, stimulated recovery from neutropenia, and induced mobilization of hematopoietic progenitor cells. In addition, the administration of G-CSF resulted in maturation of monocytes/macrophages. CONCLUSIONS These results support continuing efforts toward validation of the minipig as a large animal model for advanced testing of radiation countermeasures and characterization of the pathophysiology of ARS, and they suggest that the efficacy of G-CSF in improving survival after total body irradiation may involve mechanisms other than increasing the numbers of circulating granulocytes.
Collapse
Affiliation(s)
- Maria Moroni
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
|