1
|
Sethi P, Bhaskar R, Singh KK, Gupta S, Han SS, Avinash D, Abomughaid MM, Koul A, Rani B, Ghosh S, Jha NK, Sinha JK. Exploring advancements in early detection of Alzheimer's disease with molecular assays and animal models. Ageing Res Rev 2024; 100:102411. [PMID: 38986845 DOI: 10.1016/j.arr.2024.102411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Alzheimer's Disease (AD) is a challenging neurodegenerative condition, with overwhelming implications for affected individuals and healthcare systems worldwide. Animal models have played a crucial role in studying AD pathogenesis and testing therapeutic interventions. Remarkably, studies on the genetic factors affecting AD risk, such as APOE and TREM2, have provided valuable insights into disease mechanisms. Early diagnosis has emerged as a crucial factor in effective AD management, as demonstrated by clinical studies emphasizing the benefits of initiating treatment at early stages. Novel diagnostic technologies, including RNA sequencing of microglia, offer promising avenues for early detection and monitoring of AD progression. Therapeutic strategies remain to evolve, with a focus on targeting amyloid beta (Aβ) and tau pathology. Advances in animal models, such as APP-KI mice, and the advancement of anti-Aβ drugs signify progress towards more effective treatments. Therapeutically, the focus has shifted towards intricate approaches targeting multiple pathological pathways simultaneously. Strategies aimed at reducing Aβ plaque accumulation, inhibiting tau hyperphosphorylation, and modulating neuroinflammation are actively being explored, both in preclinical models and clinical trials. While challenges continue in developing validated animal models and translating preclinical findings to clinical success, the continuing efforts in understanding AD at molecular, cellular, and clinical levels offer hope for improved management and eventual prevention of this devastating disease.
Collapse
Affiliation(s)
- Paalki Sethi
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University, Jaipur, Rajsthan, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India.
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140401, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
2
|
Dubrof S, Zukaitis JG, Ahmed I, Sun W, Scheulin KM, Fang X, Jeon J, West FD, Zhao Q, Park HJ. Maternal supplementation of egg yolk modulates brain functional organization and functional outcomes of offspring. Nutr Res 2024; 131:147-158. [PMID: 39395250 DOI: 10.1016/j.nutres.2024.07.004] [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: 03/19/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 10/14/2024]
Abstract
Maternal nutrition during the perinatal stage is critical to offspring brain development. Egg yolks are a balanced and nutrient-dense food that is rich in bioactive components crucial to optimal neurodevelopment early in life. Egg consumption is often recommended to pregnant women to enhance both maternal and fetal health. We hypothesized that maternal intake of egg yolk from late gestation and throughout lactation would enhance functional organization and cognitive developmental outcomes in offspring using a pig model. Sows were fed a control diet (n = 6) or a diet containing egg yolks (n = 5, 350 mg egg yolk powder/kg BW/day, equivalent to ∼3 eggs/day for humans) from late gestation through lactation. At weaning, piglet offspring (n = 2/sow, total n = 22) underwent structural magnetic resonance imaging (MRI) and resting-state-functional MRI. Piglets underwent novel object recognition testing to assess hippocampal-dependent learning and memory. Functional MRI results demonstrated that egg yolk significantly increased functional activation in the executive network (p = 0.0343) and cerebellar network (p = 0.0253) in piglets when compared to control. Diffusion tensor imaging analysis showed that perinatal intake of egg yolks significantly increased white matter fiber length in the hippocampus (p = 0.0363) and cerebellum (p = 0.0287) in piglet offspring compared to control piglets. Furthermore, piglets from egg yolk-fed sows spent significantly more proportional frequency exploring the novel object than the familiar object in novel object recognition testing (p = 0.0370). The findings from this study support egg yolk-altered activation of specific brain networks may be associated with functional cognitive outcomes in weaning piglets.
Collapse
Affiliation(s)
- Stephanie Dubrof
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Jillien G Zukaitis
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Ishfaque Ahmed
- Department of Physics and Astronomy, University of Georgia, Athens, GA, 30602, USA
| | - Wenwu Sun
- Department of Physics and Astronomy, University of Georgia, Athens, GA, 30602, USA
| | - Kelly M Scheulin
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA; Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Xi Fang
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Julie Jeon
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Franklin D West
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA; Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Qun Zhao
- Department of Physics and Astronomy, University of Georgia, Athens, GA, 30602, USA
| | - Hea Jin Park
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
3
|
McCall MA. Pig Models in Retinal Research and Retinal Disease. Cold Spring Harb Perspect Med 2024; 14:a041296. [PMID: 37553210 PMCID: PMC10982707 DOI: 10.1101/cshperspect.a041296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
The pig has been used as a large animal model in biomedical research for many years and its use continues to increase because induced mutations phenocopy several inherited human diseases. In addition, they are continuous breeders, can be propagated by artificial insemination, have large litter sizes (on the order of mice), and can be genetically manipulated using all of the techniques that are currently available in mice. The pioneering work of Petters and colleagues set the stage for the use of the pig as a model of inherited retinal disease. In the last 10 years, the pig has become a model of choice where specific disease-causing mutations that are not phenocopied in rodents need to be studied and therapeutic approaches explored. The pig is not only used for retinal eye disease but also for the study of the cornea and lens. This review attempts to show how broad the use of the pig has become and how it has contributed to the assessment of treatments for eye disease. In the last 10 years, there have been several reviews that included the use of the pig in biomedical research (see body of the review) that included information about retinal disease. None directly discuss the use of the pig as an animal model for retinal diseases, including inherited diseases, where a single genetic mutation has been identified or for multifactorial diseases such as glaucoma and diabetic retinopathy. Although the pig is used to explore diseases of the cornea and lens, this review focuses on how and why the pig, as a large animal model, is useful for research in neural retinal disease and its treatment.
Collapse
Affiliation(s)
- Maureen A McCall
- Departments of Ophthalmology & Visual Sciences and Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, USA
| |
Collapse
|
4
|
van Onselen R, Downing TG. Uptake of β-N-methylamino-L-alanine (BMAA) into glutamate-specific synaptic vesicles: Exploring the validity of the excitotoxicity mechanism of BMAA. Neurosci Lett 2024; 821:137593. [PMID: 38103629 DOI: 10.1016/j.neulet.2023.137593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The first mechanism of toxicity proposed for the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) was excitotoxicity, and this was supported by numerous in vitro studies in which overactivation of both ionotropic and metabotropic glutamate receptors was reported. However, the excitotoxicity of BMAA is weak in comparison with other known excitotoxins and on par with that of glutamate, implying that to achieve sufficient synaptic concentrations of BMAA to cause classical in vivo excitotoxicity, BMAA must either accumulate in synapses to allow persistent glutamate receptor activation or it must be released in sufficiently high concentrations into synapses to cause the overexcitation. Since it has been shown that BMAA can be readily removed from synapses, release of high concentrations of BMAA into synapses must be shown to confirm its role as an excitotoxin in in vivo systems. This study therefore sought to evaluate the uptake of BMAA into synaptic vesicles and to determine if BMAA affects the uptake of glutamate into synaptic vesicles. There was no evidence to support uptake of BMAA into glutamate-specific synaptic vesicles but there was some indication that BMAA may affect the uptake of glutamate into synaptic vesicles. The uptake of BMAA into synaptic vesicles isolated from areas other than the cerebral cortex should be investigated before definite conclusions can be drawn about the role of BMAA as an excitotoxin.
Collapse
Affiliation(s)
- Rianita van Onselen
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa; Department of Biochemistry and Microbiology, Nelson Mandela University, Gqeberha, South Africa
| | - Tim G Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, Gqeberha, South Africa.
| |
Collapse
|
5
|
Andreis FR, Metcalfe B, Janjua TAM, Fazan VPS, Jensen W, Meijs S, Nielsen TGNDS. Morphology and morphometry of the ulnar nerve in the forelimb of pigs. Anat Histol Embryol 2024; 53:e12972. [PMID: 37715494 DOI: 10.1111/ahe.12972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
The knowledge of the morphology and morphometry of peripheral nerves is essential for developing neural interfaces and understanding nerve regeneration in basic and applied research. Currently, the most adopted animal model is the rat, even though recent studies have suggested that the neuroanatomy of large animal models is more comparable to humans. The present knowledge of the morphological structure of large animal models is limited; therefore, the present study aims to describe the morphological characteristics of the Ulnar Nerve (UN) in pigs. UN cross-sections were taken from seven Danish landrace pigs at three distinct locations: distal UN, proximal UN and at the dorsal cutaneous branch of the UN (DCBUN). The nerve diameter, fascicle diameter and number, number of fibres and fibre size were quantified. The UN diameter was larger in the proximal section compared to the distal segment and the DCBUN. The proximal branch also had a more significant number of fascicles (median: 15) than the distal (median: 10) and the DCBUN (median: 11) segments. Additionally, the mean fascicle diameter was smaller at the DCBUN (mean: 165 μm) than at the distal (mean: 197 μm) and proximal (mean: 199 μm) segments of the UN. Detailed knowledge of the microscopical structure of the UN in pigs is critical for further studies investigating neural interface designs and computational models of the peripheral nervous system.
Collapse
Affiliation(s)
- Felipe Rettore Andreis
- Department of Health Science and Technology, Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark
| | - Benjamin Metcalfe
- Bath Institute for the Augmented Human, University of Bath, Bath, UK
| | - Taha Al Muhammadee Janjua
- Department of Health Science and Technology, Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark
| | - Valéria Paula Sassoli Fazan
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Winnie Jensen
- Department of Health Science and Technology, Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark
| | - Suzan Meijs
- Department of Health Science and Technology, Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
6
|
Uh K, Monarch K, Reese ED, Rodriguez K, Yoon J, Spate LD, Samuel MS, Koh S, Chen PR, Jarome TJ, Allen TA, Prather RS, Lee K. Impaired Skeletal Development by Disruption of Presenilin-1 in Pigs and Generation of Novel Pig Models for Alzheimer's Disease. J Alzheimers Dis 2024; 101:445-461. [PMID: 39177593 PMCID: PMC11492100 DOI: 10.3233/jad-231297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 08/24/2024]
Abstract
Background Presenilin 1 (PSEN1) is one of the genes linked to the prevalence of early onset Alzheimer's disease. In mice, inactivation of Psen1 leads to developmental defects, including vertebral malformation and neural development. However, little is known about the role of PSEN1 during the development in other species. Objective To investigate the role of PSEN1 in vertebral development and the pathogenic mechanism of neurodegeneration using a pig model. Methods CRISPR/Cas9 system was used to generate pigs with different mutations flanking exon 9 of PSEN1, including those with a deleted exon 9 (Δexon9). Vertebral malformations in PSEN1 mutant pigs were examined by X-ray, micro-CT and micro-MRI. Neuronal cells from the brains of PSEN1 mutant pigs were analyzed by immunoflourescence, followed by image analysis including morphometric evaluation via image J and 3D reconstruction. Results Pigs with a PSEN1 null mutation (Δexon9-12) died shortly after birth and had significant axial skeletal defects, whereas pigs carrying at least one Δexon9 allele developed normally and remained healthy. Effects of the null mutation on abnormal skeletal development were also observed in fetuses at day 40 of gestation. Abnormal distribution of astrocytes and microglia in the brain was detected in two PSEN1 mutant pigs examined compared to age-matched control pigs. The founder pigs were bred to establish and age PSEN1ΔE9/+ pigs to study their relevance to clinical Alzheimer's diseases. Conclusions PSEN1 has a critical role for normal vertebral development and PSEN1 mutant pigs serves as novel resources to study Alzheimer's disease.
Collapse
Affiliation(s)
- Kyungjun Uh
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do, Republic of Korea
| | - Kaylynn Monarch
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Emily D. Reese
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | | | - Junchul Yoon
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Lee D. Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Melissa S. Samuel
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Sehwon Koh
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| | - Paula R. Chen
- United States Department of Agriculture-Agricultural Research Service, Plant Genetics Research Unit, Columbia, MO, USA
| | - Timothy J. Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Timothy A. Allen
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, USA
- Department of Environmental & Occupational Health, Robert Stempel College of Public Health, Florida International University, Miami, FL, USA
| | - Randall S. Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Kiho Lee
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
7
|
Strawn M, Safranski TJ, Behura SK. Does DNA methylation in the fetal brain leave an epigenetic memory in the blood? Gene 2023; 887:147788. [PMID: 37696423 DOI: 10.1016/j.gene.2023.147788] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/23/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Epigenetic memory is an emerging concept that refers to the process in which epigenetic changes occurring early-in life can lead to long-term programs of gene regulation in time and space. By leveraging neural network regression modeling of DNA methylation data in pigs, we show that specific methylations in the adult blood can reliably predict methylation changes that occurred in the fetal brain. Genes associated with these methylations represented known markers of specific cell types of blood including bone marrow hematopoietic progenitor cells, and ependymal and oligodendrocyte cells of brain. This suggested that methylation changes that occurred in the developing brain were maintained as an epigenetic memory in the blood through the adult life.
Collapse
Affiliation(s)
- Monica Strawn
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Timothy J Safranski
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, United States; MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, United States; Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
8
|
Kelty TJ, Taylor CL, Wieschhaus NE, Thorne PK, Amin AR, Mueller CM, Olver TD, Tharp DL, Emter CA, Caulk AW, Rector RS. Western diet-induced obesity results in brain mitochondrial dysfunction in female Ossabaw swine. Front Mol Neurosci 2023; 16:1320879. [PMID: 38163062 PMCID: PMC10755880 DOI: 10.3389/fnmol.2023.1320879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Diet-induced obesity is implicated in the development of a variety of neurodegenerative disorders. Concurrently, the loss of mitochondrial Complex I protein or function is emerging as a key phenotype across an array of neurodegenerative disorders. Therefore, the objective of this study was to determine if Western diet (WD) feeding in swine [carbohydrate = 40.8% kCal (17.8% of total calories from high fructose corn syrup), protein = 16.2% kcal, fat = 42.9% kCal, and 2% cholesterol] would result in Complex I syndrome pathology. To characterize the effects of WD-induced obesity on brain mitochondria in swine, high resolution respirometry measurements from isolated brain mitochondria, oxidative phosphorylation Complex expression, and indices of oxidative stress and mitochondrial biogenesis were assessed in female Ossabaw swine fed a WD for 6-months. In line with Complex I syndrome, WD feeding severely reduced State 3 Complex I, State 3 Complex I and II, and uncoupled mitochondrial respiration in the hippocampus and prefrontal cortex (PFC). State 3 Complex I mitochondrial respiration in the PFC inversely correlated with serum total cholesterol. WD feeding also significantly reduced protein expression of oxidative phosphorylation Complexes I-V in the PFC. WD feeding significantly increased markers of antioxidant defense and mitochondrial biogenesis in the hippocampi and PFC. These data suggest WD-induced obesity may contribute to Complex I syndrome pathology by increasing oxidative stress, decreasing oxidative phosphorylation Complex protein expression, and reducing brain mitochondrial respiration. Furthermore, these findings provide mechanistic insight into the clinical link between obesity and mitochondrial Complex I related neurodegenerative disorders.
Collapse
Affiliation(s)
- Taylor J. Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Chris L. Taylor
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - Pamela K. Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Amira R. Amin
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Christina M. Mueller
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - T. Dylan Olver
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
9
|
Banbury C, Harris G, Clancy M, Blanch RJ, Rickard JJS, Goldberg Oppenheimer P. Window into the mind: Advanced handheld spectroscopic eye-safe technology for point-of-care neurodiagnostic. SCIENCE ADVANCES 2023; 9:eadg5431. [PMID: 37967190 PMCID: PMC10651125 DOI: 10.1126/sciadv.adg5431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 10/19/2023] [Indexed: 11/17/2023]
Abstract
Traumatic brain injury (TBI), a major cause of morbidity and mortality worldwide, is hard to diagnose at the point of care with patients often exhibiting no clinical symptoms. There is an urgent need for rapid point-of-care diagnostics to enable timely intervention. We have developed a technology for rapid acquisition of molecular fingerprints of TBI biochemistry to safely measure proxies for cerebral injury through the eye, providing a path toward noninvasive point-of-care neurodiagnostics using simultaneous Raman spectroscopy and fundus imaging of the neuroretina. Detection of endogenous neuromarkers in porcine eyes' posterior revealed enhancement of high-wave number bands, clearly distinguishing TBI and healthy cohorts, classified via artificial neural network algorithm for automated data interpretation. Clinically, translating into reduced specialist support, this markedly improves the speed of diagnosis. Designed as a hand-held cost-effective technology, it can allow clinicians to rapidly assess TBI at the point of care and identify long-term changes in brain biochemistry in acute or chronic neurodiseases.
Collapse
Affiliation(s)
- Carl Banbury
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Georgia Harris
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Michael Clancy
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Ministry of Justice, 102 Petty France, Westminster, London, UK
| | - Richard J. Blanch
- Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, Robert Aiken Institute for Clinical Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Department of Ophthalmology, Queen Elizabeth Hospital Birmingham, UHB NHS Foundation Trust, West Midlands, UK
| | | | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Healthcare Technologies Institute, Institute of Translational Medicine, Mindelsohn Way, Birmingham, B15 2TH, UK
| |
Collapse
|
10
|
Allen LM, Murphy DA, Roldan V, Moussa MN, Draper A, Delgado A, Aguiar M, Capote MA, Jarome TJJ, Lee K, Mattfeld AT, Prather R, Allen TA. Testing spatial working memory in pigs using an automated T-maze. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad010. [PMID: 38596242 PMCID: PMC10913826 DOI: 10.1093/oons/kvad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 04/11/2024]
Abstract
Pigs are an important large animal model for translational clinical research but underutilized in behavioral neuroscience. This is due, in part, to a lack of rigorous neurocognitive assessments for pigs. Here, we developed a new automated T-maze for pigs that takes advantage of their natural tendency to alternate. The T-maze has obvious cross-species value having served as a foundation for cognitive theories across species. The maze (17' × 13') was constructed typically and automated with flanking corridors, guillotine doors, cameras, and reward dispensers. We ran nine pigs in (1) a simple alternation task and (2) a delayed spatial alternation task. Our assessment focused on the delayed spatial alternation task which forced pigs to wait for random delays (5, 60, 120, and 240 s) and burdened spatial working memory. We also looked at self-paced trial latencies, error types, and coordinate-based video tracking. We found pigs naturally alternated but performance declined steeply across delays (R2 = 0.84). Self-paced delays had no effect on performance suggestive of an active interference model of working memory. Positional and head direction data could differentiate subsequent turns on short but not long delays. Performance levels were stable over weeks in diverse strains and sexes, and thus provide a benchmark for future neurocognitive assessments in pigs.
Collapse
Affiliation(s)
- L M Allen
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - D A Murphy
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - V Roldan
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - M N Moussa
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - A Draper
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - A Delgado
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - M Aguiar
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - M A Capote
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - T J J Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- School of Animal Science, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - K Lee
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA
| | - A T Mattfeld
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
| | - R Prather
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA
| | - T A Allen
- Cognitive Neuroscience Program, Department of Psychology, Florida International University, Miami, FL, 33199, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
11
|
Pavlichenko M, Lafrenaye AD. The Central Fluid Percussion Brain Injury in a Gyrencephalic Pig Brain: Scalable Diffuse Injury and Tissue Viability for Glial Cell Immunolabeling following Long-Term Refrigerated Storage. Biomedicines 2023; 11:1682. [PMID: 37371777 PMCID: PMC10295711 DOI: 10.3390/biomedicines11061682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people annually; however, our knowledge of the diffuse pathologies associated with TBI is limited. As diffuse pathologies, including axonal injury and neuroinflammatory changes, are difficult to visualize in the clinical population, animal models are used. In the current study, we used the central fluid percussion injury (CFPI) model in a micro pig to study the potential scalability of these diffuse pathologies in a gyrencephalic brain of a species with inflammatory systems very similar to humans. We found that both axonal injury and microglia activation within the thalamus and corpus callosum are positively correlated with the weight-normalized pressure pulse, while subtle changes in blood gas and mean arterial blood pressure are not. We also found that the majority of tissue generated up to 10 years previously is viable for immunofluorescent labeling after long-term refrigeration storage. This study indicates that a micro pig CFPI model could allow for specific investigations of various degrees of diffuse pathological burdens following TBI.
Collapse
Affiliation(s)
- Mark Pavlichenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
| | - Audrey D. Lafrenaye
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
- Richmond Veterans Affairs Medical Center, Richmond, VA 23249-4915, USA
| |
Collapse
|
12
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Domínguez-Oliva A, Hernández-Ávalos I, Martínez-Burnes J, Olmos-Hernández A, Verduzco-Mendoza A, Mota-Rojas D. The Importance of Animal Models in Biomedical Research: Current Insights and Applications. Animals (Basel) 2023; 13:ani13071223. [PMID: 37048478 PMCID: PMC10093480 DOI: 10.3390/ani13071223] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/19/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Animal research is considered a key element in advance of biomedical science. Although its use is controversial and raises ethical challenges, the contribution of animal models in medicine is essential for understanding the physiopathology and novel treatment alternatives for several animal and human diseases. Current pandemics’ pathology, such as the 2019 Coronavirus disease, has been studied in primate, rodent, and porcine models to recognize infection routes and develop therapeutic protocols. Worldwide issues such as diabetes, obesity, neurological disorders, pain, rehabilitation medicine, and surgical techniques require studying the process in different animal species before testing them on humans. Due to their relevance, this article aims to discuss the importance of animal models in diverse lines of biomedical research by analyzing the contributions of the various species utilized in science over the past five years about key topics concerning human and animal health.
Collapse
Affiliation(s)
- Adriana Domínguez-Oliva
- Master’s Program in Agricultural and Livestock Sciences [Maestría en Ciencias Agropecuarias], Xochimilco Campus, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán 54714, Mexico
| | - Julio Martínez-Burnes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Victoria City 87000, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
- Correspondence:
| |
Collapse
|
14
|
Knoernschild K, Johnson HJ, Schroeder KE, Swier VJ, White KA, Sato TS, Rogers CS, Weimer JM, Sieren JC. Magnetic resonance brain volumetry biomarkers of CLN2 Batten disease identified with miniswine model. Sci Rep 2023; 13:5146. [PMID: 36991106 PMCID: PMC10060411 DOI: 10.1038/s41598-023-32071-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease (Batten disease) is a rare pediatric disease, with symptom development leading to clinical diagnosis. Early diagnosis and effective tracking of disease progression are required for treatment. We hypothesize that brain volumetry is valuable in identifying CLN2 disease at an early stage and tracking disease progression in a genetically modified miniswine model. CLN2R208X/R208X miniswine and wild type controls were evaluated at 12- and 17-months of age, correlating to early and late stages of disease progression. Magnetic resonance imaging (MRI) T1- and T2-weighted data were acquired. Total intercranial, gray matter, cerebrospinal fluid, white matter, caudate, putamen, and ventricle volumes were calculated and expressed as proportions of the intracranial volume. The brain regions were compared between timepoints and cohorts using Gardner-Altman plots, mean differences, and confidence intervals. At an early stage of disease, the total intracranial volume (- 9.06 cm3), gray matter (- 4.37% 95 CI - 7.41; - 1.83), caudate (- 0.16%, 95 CI - 0.24; - 0.08) and putamen (- 0.11% 95 CI - 0.23; - 0.02) were all notably smaller in CLN2R208X/R208X miniswines versus WT, while cerebrospinal fluid was larger (+ 3.42%, 95 CI 2.54; 6.18). As the disease progressed to a later stage, the difference between the gray matter (- 8.27%, 95 CI - 10.1; - 5.56) and cerebrospinal fluid (+ 6.88%, 95 CI 4.31; 8.51) continued to become more pronounced, while others remained stable. MRI brain volumetry in this miniswine model of CLN2 disease is sensitive to early disease detection and longitudinal change monitoring, providing a valuable tool for pre-clinical treatment development and evaluation.
Collapse
Affiliation(s)
- Kevin Knoernschild
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kimberly E Schroeder
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | - Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Takashi S Sato
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | | | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
da Silva BPM, Fanalli SL, Gomes JD, de Almeida VV, Fukumasu H, Freitas FAO, Moreira GCM, Silva-Vignato B, Reecy JM, Koltes JE, Koltes D, de Carvalho Balieiro JC, de Alencar SM, da Silva JPM, Coutinho LL, Afonso J, Regitano LCDA, Mourão GB, Luchiari Filho A, Cesar ASM. Brain fatty acid and transcriptome profiles of pig fed diets with different levels of soybean oil. BMC Genomics 2023; 24:91. [PMID: 36855067 PMCID: PMC9976441 DOI: 10.1186/s12864-023-09188-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/15/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND The high similarity in anatomical and neurophysiological processes between pigs and humans make pigs an excellent model for metabolic diseases and neurological disorders. Lipids are essential for brain structure and function, and the polyunsaturated fatty acids (PUFA) have anti-inflammatory and positive effects against cognitive dysfunction in neurodegenerative diseases. Nutrigenomics studies involving pigs and fatty acids (FA) may help us in better understanding important biological processes. In this study, the main goal was to evaluate the effect of different levels of dietary soybean oil on the lipid profile and transcriptome in pigs' brain tissue. RESULTS Thirty-six male Large White pigs were used in a 98-day study using two experimental diets corn-soybean meal diet containing 1.5% soybean oil (SOY1.5) and corn-soybean meal diet containing 3.0% soybean oil (SOY3.0). No differences were found for the brain total lipid content and FA profile between the different levels of soybean oil. For differential expression analysis, using the DESeq2 statistical package, a total of 34 differentially expressed genes (DEG, FDR-corrected p-value < 0.05) were identified. Of these 34 DEG, 25 are known-genes, of which 11 were up-regulated (log2 fold change ranging from + 0.25 to + 2.93) and 14 were down-regulated (log2 fold change ranging from - 3.43 to -0.36) for the SOY1.5 group compared to SOY3.0. For the functional enrichment analysis performed using MetaCore with the 34 DEG, four pathway maps were identified (p-value < 0.05), related to the ALOX15B (log2 fold change - 1.489), CALB1 (log2 fold change - 3.431) and CAST (log2 fold change + 0.421) genes. A "calcium transport" network (p-value = 2.303e-2), related to the CAST and CALB1 genes, was also identified. CONCLUSION The results found in this study contribute to understanding the pathways and networks associated with processes involved in intracellular calcium, lipid metabolism, and oxidative processes in the brain tissue. Moreover, these results may help a better comprehension of the modulating effects of soybean oil and its FA composition on processes and diseases affecting the brain tissue.
Collapse
Affiliation(s)
- Bruna Pereira Martins da Silva
- grid.11899.380000 0004 1937 0722Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Simara Larissa Fanalli
- grid.11899.380000 0004 1937 0722Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Julia Dezen Gomes
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Vivian Vezzoni de Almeida
- grid.411195.90000 0001 2192 5801College of Veterinary Medicine and Animal Science, Federal University of Goiás, Goiânia, Goiás Brazil
| | - Heidge Fukumasu
- grid.11899.380000 0004 1937 0722Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Felipe André Oliveira Freitas
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | | | - Bárbara Silva-Vignato
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - James Mark Reecy
- grid.34421.300000 0004 1936 7312College of Agriculture and Life Sciences, Iowa State University, Ames, IA USA
| | - James Eugene Koltes
- grid.34421.300000 0004 1936 7312College of Agriculture and Life Sciences, Iowa State University, Ames, IA USA
| | - Dawn Koltes
- grid.34421.300000 0004 1936 7312College of Agriculture and Life Sciences, Iowa State University, Ames, IA USA
| | - Júlio Cesar de Carvalho Balieiro
- grid.11899.380000 0004 1937 0722School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Severino Matias de Alencar
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Julia Pereira Martins da Silva
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Luiz Lehmann Coutinho
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Juliana Afonso
- grid.460200.00000 0004 0541 873XEmbrapa Pecuária Sudeste, São Carlos, São Paulo, Brazil
| | | | - Gerson Barreto Mourão
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Albino Luchiari Filho
- grid.11899.380000 0004 1937 0722Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Aline Silva Mello Cesar
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil. .,Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil.
| |
Collapse
|
16
|
Limborska SA, Filippenkov IB. Special Issue "Genomics of Stroke" 2022. Genes (Basel) 2023; 14:514. [PMID: 36833441 PMCID: PMC9956966 DOI: 10.3390/genes14020514] [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: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Stroke is one of the greatest medical threats to human health and quality of life in modern society [...].
Collapse
Affiliation(s)
| | - Ivan B. Filippenkov
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia
| |
Collapse
|
17
|
Pupillary Light Response Deficits in 4-Week-Old Piglets and Adolescent Children after Low-Velocity Head Rotations and Sports-Related Concussions. Biomedicines 2023; 11:biomedicines11020587. [PMID: 36831121 PMCID: PMC9952885 DOI: 10.3390/biomedicines11020587] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Neurological disorders and traumatic brain injury (TBI) are among the leading causes of death and disability. The pupillary light reflex (PLR) is an emerging diagnostic tool for concussion in humans. We compared PLR obtained with a commercially available pupillometer in the 4 week old piglet model of the adolescent brain subject to rapid nonimpact head rotation (RNR), and in human adolescents with and without sports-related concussion (SRC). The 95% PLR reference ranges (RR, for maximum and minimum pupil diameter, latency, and average and peak constriction velocities) were established in healthy piglets (N = 13), and response reliability was validated in nine additional healthy piglets. PLR assessments were obtained in female piglets allocated to anesthetized sham (N = 10), single (sRNR, N = 13), and repeated (rRNR, N = 14) sagittal low-velocity RNR at pre-injury, as well as days 1, 4, and 7 post injury, and evaluated against RRs. In parallel, we established human PLR RRs in healthy adolescents (both sexes, N = 167) and compared healthy PLR to values obtained <28 days from a SRC (N = 177). In piglets, maximum and minimum diameter deficits were greater in rRNR than sRNR. Alterations peaked on day 1 post sRNR and rRNR, and remained altered at day 4 and 7. In SRC adolescents, the proportion of adolescents within the RR was significantly lower for maximum pupil diameter only (85.8%). We show that PLR deficits may persist in humans and piglets after low-velocity head rotations. Differences in timing of assessment after injury, developmental response to injury, and the number and magnitude of impacts may contribute to the differences observed between species. We conclude that PLR is a feasible, quantifiable involuntary physiological metric of neurological dysfunction in pigs, as well as humans. Healthy PLR porcine and human reference ranges established can be used for neurofunctional assessments after TBI or hypoxic exposures (e.g., stroke, apnea, or cardiac arrest).
Collapse
|
18
|
Li K, Cardoso C, Moctezuma-Ramirez A, Elgalad A, Perin E. Evaluation of large animal models for preclinical studies of heart failure with preserved ejection fraction using clinical score systems. Front Cardiovasc Med 2023; 10:1099453. [PMID: 37034319 PMCID: PMC10076838 DOI: 10.3389/fcvm.2023.1099453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a complex, heterogeneous spectrum of pathologic features combined with average left ventricular volume and diastolic dysfunction. HFpEF is a significant public health problem associated with high morbidity and mortality rates. Currently, effective treatments for HFpEF represent the greatest unmet need in cardiovascular medicine. A lack of an efficient preclinical model has hampered the development of new devices and medications for HFpEF. Because large animal models have similar physiologic traits as humans and appropriate organ sizes, they are the best option for limiting practical constraints. HFpEF is a highly integrated, multiorgan, systemic disorder requiring a multipronged investigative approach. Here, we review the large animal models of HFpEF reported to date and describe the methods that have been used to create HFpEF, including surgery-induced pressure overloading, medicine-induced pressure overloading, and diet-induced metabolic syndrome. In addition, for the first time to our knowledge, we use two established clinical HFpEF algorithms (HFA-PEFF and H2FPEF scores) to evaluate the currently available large animal models. We also discuss new technologies, such as continuous remote pressure monitors and inflatable aortic cuffs, as well as how the models could be improved. Based on current progress and our own experience, we believe an efficient large animal model of HFpEF should simultaneously encompass multiple pathophysiologic factors, along with multiorgan dysfunction. This could be fully evaluated through available methods (imaging, blood work). Although many models have been studied, only a few studies completely meet clinical score standards. Therefore, it is critical to address the deficiencies of each model and incorporate novel techniques to establish a more reliable model, which will help facilitate the understanding of HFpEF mechanisms and the development of a treatment.
Collapse
Affiliation(s)
- Ke Li
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Cristiano Cardoso
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Angel Moctezuma-Ramirez
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
| | - Abdelmotagaly Elgalad
- Center for Preclinical Cardiovascular Research, The Texas Heart Institute, Houston, TX, United States
- Correspondence: Abdelmotagaly Elgalad
| | - Emerson Perin
- Center for Clinical Research, The Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
19
|
Real CC, Binda KH, Thomsen MB, Lillethorup TP, Brooks DJ, Landau AM. Selecting the Best Animal Model of Parkinson's Disease for Your Research Purpose: Insight from in vivo PET Imaging Studies. Curr Neuropharmacol 2023; 21:1241-1272. [PMID: 36797611 PMCID: PMC10286593 DOI: 10.2174/1570159x21666230216101659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 02/18/2023] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative multisystem disorder leading to motor and non-motor symptoms in millions of individuals. Despite intense research, there is still no cure, and early disease biomarkers are lacking. Animal models of PD have been inspired by basic elements of its pathogenesis, such as dopamine dysfunction, alpha-synuclein accumulation, neuroinflammation and disruption of protein degradation, and these have been crucial for a deeper understanding of the mechanisms of pathology, the identification of biomarkers, and evaluation of novel therapies. Imaging biomarkers are non-invasive tools to assess disease progression and response to therapies; their discovery and validation have been an active field of translational research. Here, we highlight different considerations of animal models of PD that can be applied to future research, in terms of their suitability to answer different research questions. We provide the reader with important considerations of the best choice of model to use based on the disease features of each model, including issues related to different species. In addition, positron emission tomography studies conducted in PD animal models in the last 5 years are presented. With a variety of different species, interventions and genetic information, the choice of the most appropriate model to answer research questions can be daunting, especially since no single model recapitulates all aspects of this complex disorder. Appropriate animal models in conjunction with in vivo molecular imaging tools, if selected properly, can be a powerful combination for the assessment of novel therapies and developing tools for early diagnosis.
Collapse
Affiliation(s)
- Caroline Cristiano Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karina Henrique Binda
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Majken Borup Thomsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David James Brooks
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle, Upon Tyne, UK
| | - Anne Marlene Landau
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
Zhu F, Wang H, Li L, Bragin A, Cao D, Cheng Y. Intracranial electrophysiological recordings on a swine model of mesial temporal lobe epilepsy. Front Neurol 2023; 14:1077702. [PMID: 37139062 PMCID: PMC10150775 DOI: 10.3389/fneur.2023.1077702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/20/2023] [Indexed: 05/05/2023] Open
Abstract
Objective To test the feasibility and reliability of intracranial electrophysiological recordings in an acute status epilepticus model on laboratory swine. Method Intrahippocampal injection of kainic acid (KA) was performed on 17 male Bama pigs (Sus scrofa domestica) weighing between 25 and 35 kg. Two stereoelectroencephalography (SEEG) electrodes with a total of 16 channels were implanted bilaterally along the sensorimotor cortex to the hippocampus. Brain electrical activity was recorded 2 h daily for 9-28 days. Three KA dosages were tested to evaluate the quantities capable of evoking status epilepticus. Local field potentials (LFPs) were recorded and compared before and after the KA injection. We quantified the epileptic patterns, including the interictal spikes, seizures, and high-frequency oscillations (HFOs), up to 4 weeks after the KA injection. Test-retest reliability using intraclass correlation coefficients (ICCs) were performed on interictal HFO rates to evaluate the recording stability of this model. Results The KA dosage test suggested that a 10 μl (1.0 μg/μl) intrahippocampal injection could successfully evoke status epilepticus lasting from 4 to 12 h. At this dosage, eight pigs (50% of total) had prolonged epileptic events (tonic-chronic seizures + interictal spikes n = 5, interictal spikes alone n = 3) in the later 4 weeks of the video-SEEG recording period. Four pigs (25% of total) had no epileptic activities, and another four (25%) had lost the cap or did not complete the experiments. Animals that showed epileptiform events were grouped as E + (n = 8) and the four animals showing no signs of epileptic events were grouped as E- (n = 4). A total of 46 electrophysiological seizures were captured in the 4-week post-KA period from 4 E + animals, with the earliest onset on day 9. The seizure durations ranged from 12 to 45 s. A significant increase of hippocampal HFOs rate (num/min) was observed in the E+ group during the post-KA period (weeks 1, 2,4, p < 0.05) compared to the baseline. But the E-showed no change or a decrease (in week 2, p = 0.43) compared to their baseline rate. The between-group comparison showed much higher HFO rates in E + vs. E - (F = 35, p < 0.01). The high ICC value [ICC (1, k) = 0.81, p < 0.05] quantified from the HFO rate suggested that this model had a stable measurement of HFOs during the four-week post-KA periods. Significance This study measured intracranial electrophysiological activity in a swine model of KA-induced mesial temporal lobe epilepsy (mTLE). Using the clinical SEEG electrode, we distinguished abnormal EEG patterns in the swine brain. The high test-retest reliability of HFO rates in the post-KA period suggests the utility of this model for studying mechanisms of epileptogenesis. The use of swine may provide satisfactory translational value for clinical epilepsy research.
Collapse
Affiliation(s)
- Fengjun Zhu
- Department of Neurosurgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
- Department of Neurosurgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, United States
| | - Hanwen Wang
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, United States
| | - Lin Li
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, United States
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | - Anatol Bragin
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, United States
| | - Dezhi Cao
- Department of Neurosurgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Dezhi Cao,
| | - Yuan Cheng
- Department of Neurosurgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Yuan Cheng,
| |
Collapse
|
21
|
Mariager T, Bjarkam C, Nielsen H, Bodilsen J. Experimental animal models for brain abscess: a systematic review. Br J Neurosurg 2022:1-8. [PMID: 36579498 DOI: 10.1080/02688697.2022.2160865] [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: 12/09/2020] [Revised: 07/04/2022] [Accepted: 10/18/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Brain abscess (BA) is a rare, but severe infection and experimental BA animal models may prove crucial for advances in treatment. This review describes the development of experimental BA models and the clinical advances obtained from these, in a historical perspective. MATERIAL AND METHODS Experimental BA studies from inception until June 15, 2022, were included by searching the PubMed and Embase databases. Inclusion required the use of an experimental BA animal model. Non-bacterial BA models, in vitro studies, veterinarian case-reports, and articles written in non-English language were excluded. Bias was not systematically assessed, and the review was not registered at the PROSPERO. RESULTS 79 studies were included. The majority of animal BA models have been based on small rodents using Staphylococcus aureus. The models have delineated the natural development of BA and provided detailed descriptions of the histopathological characteristics consisting of a necrotic centre surrounded by layers of inflammatory cells and fibroblasts encapsulated by a dense collagenous layer. Radiological studies of animal BA have been shown to correlate with the corresponding stages of human BA in both computed tomography and magnetic resonance imaging and may guide diagnosis as well as the timing of neurosurgical intervention. Moreover, pharmacokinetic studies of the intracavitary penetration of various antimicrobials have helped inform medical treatment of BA. Other studies have examined the diverse effects of corticosteroids including decreased cerebral oedema, intracranial pressure, and intracavitary drug concentration, whereas concerns on decreased or weakened capsule formation could not be confirmed. Finally, studies on the immunological response to BA have highlighted potential future immunomodulatory targets. CONCLUSIONS Animal models have been vital for improvements in the management of BA. Experimental BA models resembling human disease including polymicrobial infection by oral cavity flora in large animals are needed.
Collapse
Affiliation(s)
- Theis Mariager
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Carsten Bjarkam
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Nielsen
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
| | - Jacob Bodilsen
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
22
|
Fedulova L, Vasilevskaya E, Tikhonova O, Kazieva L, Tolmacheva G, Makarenko A. Proteomic Markers in the Muscles and Brain of Pigs Recovered from Hemorrhagic Stroke. Genes (Basel) 2022; 13:genes13122204. [PMID: 36553471 PMCID: PMC9777686 DOI: 10.3390/genes13122204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
(1) Background: Stroke is the leading cause of serious long-term disability. Walking dysfunction and paresis of the upper extremities occurs in more than 80% of people who have had a stroke. (2) Methods: We studied post-genomic markers in biosamples of muscle and brain tissue from animals that underwent intracerebral hematoma and recovered after 42 days. Our purpose was to understand the biological mechanisms associated with recovery from hemorrhagic stroke. We analyzed the peptides formed after trypsinolysis of samples by HPLC-MS, and the results were processed by bioinformatics methods, including the establishment of biochemical relationships (gene to gene) using topological omics databases such as Reactome and KEGG. (3) Results: In the pig brain, unique compounds were identified which are expressed during the recovery period after traumatic injury. These are molecular factors of activated microglia, and they contribute to the functional recovery of neurons and reduce instances of hematoma, edema, and oxidative stress. Complexes of the main binding factors of the neurotrophins involved in the differentiation and survival of nerve cells were found in muscles. (4) Conclusions: A network of gene interactions has been constructed for proteins involved in the regulation of synaptic transmission, in particular presynaptic vesicular and endocytic processes. The presence of transmitters and transporters associated with stimulation of NMDA receptors at neuromuscular junctions shows the relationship between upper motor neurons and neuromuscular junctions.
Collapse
Affiliation(s)
- Liliya Fedulova
- V.M. Gorbatov Federal Research Center for Food Systems, Russian Academy of Sciences, 109316 Moscow, Russia
| | - Ekaterina Vasilevskaya
- V.M. Gorbatov Federal Research Center for Food Systems, Russian Academy of Sciences, 109316 Moscow, Russia
| | | | - Laura Kazieva
- Institute of Biomedical Chemistry, 119121 Mosow, Russia
| | - Galina Tolmacheva
- V.M. Gorbatov Federal Research Center for Food Systems, Russian Academy of Sciences, 109316 Moscow, Russia
- Correspondence: ; Tel.: +7-495-676-9511-(128)
| | - Alexandr Makarenko
- V.M. Gorbatov Federal Research Center for Food Systems, Russian Academy of Sciences, 109316 Moscow, Russia
| |
Collapse
|
23
|
Wei J, Zhang W, Li J, Jin Y, Qiu Z. Application of the transgenic pig model in biomedical research: A review. Front Cell Dev Biol 2022; 10:1031812. [PMID: 36325365 PMCID: PMC9618879 DOI: 10.3389/fcell.2022.1031812] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
The large animal model has gradually become an essential part of preclinical research studies, relating to exploring the disease pathological mechanism, genic function, pharmacy, and other subjects. Although the mouse model has already been widely accepted in clinical experiments, the need for finding an animal model with high similarity compared with a human model is urgent due to the different body functions and systems between mice and humans. The pig is an optimal choice for replacement. Therefore, enhancing the production of pigs used for models is an important part of the large animal model as well. Transgenic pigs show superiority in pig model creation because of the progress in genetic engineering. Successful cases of transgenic pig models occur in the clinical field of metabolic diseases, neurodegenerative diseases, and genetic diseases. In addition, the choice of pig breed influences the effort and efficiency of reproduction, and the mini pig has relative obvious advantages in pig model production. Indeed, pig models in these diseases provide great value in studies of their causes and treatments, especially at the genetic level. This review briefly outlines the method used to create transgenic pigs and species of producing transgenic pigs and provides an overview of their applications on different diseases and limitations for present pig model developments.
Collapse
Affiliation(s)
| | | | | | - Ye Jin
- *Correspondence: Ye Jin, ; Zhidong Qiu,
| | | |
Collapse
|
24
|
Epigenetic regulation of fetal brain development in pig. Gene 2022; 844:146823. [PMID: 35988784 DOI: 10.1016/j.gene.2022.146823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/27/2022] [Accepted: 08/15/2022] [Indexed: 02/01/2023]
Abstract
How fetal brain development is regulated at the molecular level is not well understood. Due to ethical challenges associated with research on the human fetus, large animals particularly pigs are increasingly used to study development and disorders of fetal brain. The pig fetal brain grows rapidly during the last ∼ 50 days before birth which is around day 60 (d60) of pig gestation. But what regulates the onset of accelerated growth of the brain is unknown. The current study tests the hypothesis that epigenetic alteration around d60 is involved in the onset of rapid growth of fetal brain of pig. To test this hypothesis, DNA methylation changes of fetal brain was assessed in a genome-wide manner by Enzymatic Methyl-seq (EM-seq) during two gestational periods (GP): d45 vs. d60 (GP1) and d60 vs. d90 (GP2). The cytosine-guanine (CpG) methylation data was analyzed in an integrative manner with the RNA-seq data generated from the same brain samples from our earlier study. A neural network based modeling approach was implemented to learn changes in methylation patterns of the differentially expressed genes, and then predict methylations of the brain in a genome-wide manner during rapid growth. This approach identified specific methylations that changed in a mutually informative manner during rapid growth of the fetal brain. These methylations were significantly overrepresented in specific genic as well as intergenic features including CpG islands, introns, and untranslated regions. In addition, sex-bias methylations of known single nucleotide polymorphic sites were also identified in the fetal brain ide during rapid growth.
Collapse
|
25
|
Takalloobanafshi G, Kukreja A, Hicks JW. Historical efforts to develop 99mTc-based amyloid plaque targeting radiotracers. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:963698. [PMID: 39390996 PMCID: PMC11466234 DOI: 10.3389/fnume.2022.963698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/21/2022] [Indexed: 10/12/2024]
Abstract
Imaging biomarkers have changed the way we study Alzheimer's disease and related dementias, develop new therapeutics to treat the disease, and stratify patient populations in clinical trials. With respect to protein aggregates comprised of amyloid-β plaques and tau neurofibrillary tangles, Positron Emission Tomography (PET) has become the gold standard imaging modality for quantitative visualization. Due to high infrastructural costs, the availability of PET remains limited to large urban areas within high income nations. This limits access to leading edge medical imaging, and potentially access to new treatments, by millions of rural and remote residents in those regions as well as billions of people in middle- and low-income countries. Single Photon Emission Computed Tomography (SPECT) is a more widely available imaging alternative with lower infrastructural costs and decades of familiarity amongst nuclear medicine professionals. Recent technological advances have closed the gap in spatial resolution and quantitation between SPECT and PET. If effective SPECT radiotracers were available to visualize amyloid-β plaques, geographic barriers to imaging could be circumvented. In this review, we will discuss past efforts to develop SPECT radiotracers targeting amyloid-β plaques which incorporate the most used radionuclide in nuclear medicine: technetium-99m (99mTc; t 1/2 = 6.01 h; γ = 140 keV). While reviewing the various chemical scaffolds and chelates employed, the focus will be upon the impact to the pharmacological properties of putative 99mTc-based amyloid-targeting radiotracers.
Collapse
Affiliation(s)
- Ghazaleh Takalloobanafshi
- Department of Chemistry, Western University, London, ON, Canada
- Cyclotron and Radiochemistry Facility, Lawson Health Research Institute, London, ON, Canada
| | - Aditi Kukreja
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Justin W. Hicks
- Cyclotron and Radiochemistry Facility, Lawson Health Research Institute, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
- Saint Joseph's Health Care London, London, ON, Canada
| |
Collapse
|
26
|
Regional Characterization of the Gottingen Minipig Brain by [18 F]FDG Dynamic Pet Modeling. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00739-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Abstract
Purpose
To determine the best kinetic model to be applied on dynamic brain [18 F]FDG PET images by characterizing the regional brain glucose metabolism of normal Göttingen minipigs.
Methods
Nine Göttingen minipigs were scanned with a clinical PET/CT tomograph, starting from the injection of an intravenous bolus of [18 F]FDG, for about 25 min. Dynamic images were reconstructed and nine brain regions of interest (ROI), plus a vascular region, were defined and time-activity curves (TAC) were determined.
Three kinetic models were considered for fitting with experimental TACs: one-tissue compartment model 1TC, two-tissue irreversible compartment model 2TCi and two-tissue reversible model 2TC. Akaike Information Criterion was considered to evaluate the goodness of each model fitting. Regional and global kinetic parameter values were evaluated, in addition to the partition coefficient, net influx rate and retention index (RI).
Results
Both 2TCi and 2TC models turned out to be good choices for the next analysis. Parameter values were very similar between the different brain regions, with similar values to when the brain as a whole is considered (kinetic parameters mean values, from 2TCi model: K1 = 1.0 ml/g/min, k2 = 0.49 min− 1, k3 = 0.034 min− 1, K1/k2 = 2.14ml/g, Ki =0.069 ml/g/min; from 2TC model: K1 = 1.10 ml/g/min, k2 = 0.54 min− 1, k3 = 0.058 min− 1, k4 = 0.039 min− 1, K1/k2 = 2.18 ml/g, Ki = 0.10 ml/g/min; RI mean ± sd: 0.147 ± 0.037 min− 1), with the exception of the cerebellum (mean values from the 2TCi model: K1 = 0.52 ml/g/min, k2 = 0.56 min− 1, k3 = 0.025 min− 1, K1/k2 = 0.98ml/g, Ki=0.022 ml/g/min; from 2TC model: K1 = 0.54 ml/g/min, k2 = 0.61 min− 1, k3 = 0.044 min− 1, k4 = 0.038 min− 1, K1/k2 = 0.95ml/g, Ki=0.032 ml/g/min; RI mean ± sd: 0.071 ± 0.018 min− 1).
Conclusion
The two-tissue model is able to describe the regional brain metabolism in Göttingen minipigs. Compared to the 2TCi model, in the 2TC model the k4 micro-parameter was also evaluated. This led to adjustments of the other microparameters, especially k3 and consequently the net influx rate Ki. For healthy minipigs, the glucose metabolism was similar in all of the brain regions analyzed, with the exception of the cerebellum, where the FDG uptake was lower.
Collapse
|
27
|
Identification of Body Size Determination Related Candidate Genes in Domestic Pig Using Genome-Wide Selection Signal Analysis. Animals (Basel) 2022; 12:ani12141839. [PMID: 35883386 PMCID: PMC9312078 DOI: 10.3390/ani12141839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 01/03/2023] Open
Abstract
This study aimed to identify the genes related to the body size of pigs by conducting genome-wide selection analysis (GWSA). We performed a GWSA scan on 50 pigs belonging to four small-bodied pig populations (Diannan small-eared pig, Bama Xiang pig, Wuzhishan pig, and Jeju black pig from South Korea) and 124 large-bodied pigs. We used the genetic parameters of the pairwise fixation index (FST) and π ratio (case/control) to screen candidate genome regions and genes related to body size. The results revealed 47,339,509 high-quality SNPs obtained from 174 individuals, while 280 interacting candidate regions were obtained from the top 1% signal windows of both parameters, along with 187 genes (e.g., ADCK4, AMDHD2, ASPN, ASS1, and ATP6V0C). The results of the candidate gene (CG) annotation showed that a series of CGs (e.g., MSTN, LTBP4, PDPK1, PKMYT1, ASS1, and STAT6) was enriched into the gene ontology terms. Moreover, molecular pathways, such as the PI3K-Akt, HIF-1, and AMPK signaling pathways, were verified to be related to body development. Overall, we identified a series of key genes that may be closely related to the body size of pigs, further elucidating the heredity basis of body shape determination in pigs and providing a theoretical reference for molecular breeding.
Collapse
|
28
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
29
|
Sutkus LT, Joung S, Hirvonen J, Jensen HM, Ouwehand AC, Mukherjea R, Donovan SM, Dilger RN. Influence of 2'-Fucosyllactose and Bifidobacterium longum Subspecies infantis Supplementation on Cognitive and Structural Brain Development in Young Pigs. Front Neurosci 2022; 16:860368. [PMID: 35546890 PMCID: PMC9081927 DOI: 10.3389/fnins.2022.860368] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/21/2022] [Indexed: 12/18/2022] Open
Abstract
Development of the gut-brain axis during early-life is an important contributor of brain structural and functional development. Human milk oligosaccharides and gut microbiota have potential beneficial effects on various aspects of development; however, the effects of 2′-fucosyllactose (2′-FL) and Bifidobacterium longum subsp. infantis Bi-26 (Bi-26) administration during infancy separately and combined are still not clear. Therefore, we investigated the effects of early administration of dietary 2′-FL and Bi-26 on brain structural and functional development in the young pig. From postnatal day (PND) 2–34 or 35, fifty-two intact male pigs were randomly assigned to treatment groups in a 2 × 2 factorial arrangement and provided ad libitum access to a nutritionally adequate milk replacer without or with 1.0 g of 2′-FL/L of reconstituted liquid. Pigs within each diet group were further stratified to receive a daily oral dose of glycerol stock without or with Bi-26 (109 CFU). Pigs were subjected to the novel object recognition (NOR) task from PND 27–31 to assess recognition memory and subsequently underwent magnetic resonance imaging procedures at PND 32 or 33 to assess brain macrostructure and microstructure. Pigs that received Bi-26 had smaller absolute brain volumes for 9 of 27 brain regions of interest, and smaller relative volumes for 2 regions associated with kinesthesia (P < 0.05). Synbiotic administration of 2′-FL and Bi-26 elicited interactive effects (P < 0.05) on several microstructural brain components, where dual supplementation negated the effects of each test article alone. Behavioral outcomes indicated that pigs did not express novelty preference, regardless of treatment group, demonstrating no effects of 2′-FL and Bi-26 on recognition memory when supplemented alone or in combination. Interactive effects (P < 0.05) were observed for the number of all object visits, latency to the first object visit, and number of familiar object visits. Pigs that did not receive Bi-26 supplementation exhibited less time interacting with the familiar object in total (P = 0.002) and on average (P = 0.005). In conclusion, supplementation of 2′-FL and/or Bi-26 elicited some alterations in object exploratory behaviors and macro/micro-structures of the brain, but changes in recognition memory were not observed. Specifically in brain microstructure, synbiotic administration of 2′-FL and Bi-26 appeared to negate effects observed when each dietary article was supplemented separately.
Collapse
Affiliation(s)
- Loretta T Sutkus
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Sangyun Joung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | | | - Henrik Max Jensen
- IFF R&D-Enabling Technologies, Advanced Analytical, Brabrand, Denmark
| | | | | | - Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Ryan N Dilger
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
30
|
Stirm M, Fonteyne LM, Shashikadze B, Stöckl JB, Kurome M, Keßler B, Zakhartchenko V, Kemter E, Blum H, Arnold GJ, Matiasek K, Wanke R, Wurst W, Nagashima H, Knieling F, Walter MC, Kupatt C, Fröhlich T, Klymiuk N, Blutke A, Wolf E. Pig models for Duchenne muscular dystrophy – from disease mechanisms to validation of new diagnostic and therapeutic concepts. Neuromuscul Disord 2022; 32:543-556. [DOI: 10.1016/j.nmd.2022.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
|
31
|
Epigenetic MRI: Noninvasive imaging of DNA methylation in the brain. Proc Natl Acad Sci U S A 2022; 119:e2119891119. [PMID: 35235458 PMCID: PMC8915962 DOI: 10.1073/pnas.2119891119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Dynamic epigenetic activity is a fundamental mechanism underpinning how the brain changes its function during development and aging and in response to environmental and disease stimuli. We developed a technology called epigenetic MRI (eMRI) that enables noninvasive imaging of DNA methylation in the brain, a major epigenetic mechanism. eMRI reveals strong regional differences in global DNA methylation in pig brains, a model with stronger resemblance to human brains than are rodents. Given the noninvasive nature of eMRI, our results pave the way for a DNA-methylation imaging paradigm for living human brains. We expect eMRI to enable many studies to unravel the molecular control of brain function and disease. Both neuronal and genetic mechanisms regulate brain function. While there are excellent methods to study neuronal activity in vivo, there are no nondestructive methods to measure global gene expression in living brains. Here, we present a method, epigenetic MRI (eMRI), that overcomes this limitation via direct imaging of DNA methylation, a major gene-expression regulator. eMRI exploits the methionine metabolic pathways for DNA methylation to label genomic DNA through 13C-enriched diets. A 13C magnetic resonance spectroscopic imaging method then maps the spatial distribution of labeled DNA. We validated eMRI using pigs, whose brains have stronger similarity to humans in volume and anatomy than rodents, and confirmed efficient 13C-labeling of brain DNA. We also discovered strong regional differences in global DNA methylation. Just as functional MRI measurements of regional neuronal activity have had a transformational effect on neuroscience, we expect that the eMRI signal, both as a measure of regional epigenetic activity and as a possible surrogate for regional gene expression, will enable many new investigations of human brain function, behavior, and disease.
Collapse
|
32
|
Mirkiani S, Roszko DA, O'Sullivan C, Faridi P, Hu DS, Fang D, Everaert DG, Toossi A, Konrad PE, Robinson K, Mushahwar VK. Overground gait kinematics and muscle activation patterns in the Yucatan mini pig. J Neural Eng 2022; 19. [PMID: 35172283 DOI: 10.1088/1741-2552/ac55ac] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/16/2022] [Indexed: 11/12/2022]
Abstract
Objective The objectives of this study were to assess gait biomechanics and the effect of overground walking speed on gait parameters, kinematics, and electromyographic (EMG) activity in the hindlimb muscles of Yucatan Minipigs (YMPs). Approach Nine neurologically-intact, adult YMPs were trained to walk overground in a straight line. Whole-body kinematics and EMG activity of hindlimb muscles were recorded and analyzed at 6 different speed ranges (0.4-0.59, 0.6-0.79, 0.8-0.99, 1.0-1.19, 1.2-1.39, and 1.4-1.6 m/s). A MATLAB program was developed to detect strides and gait events automatically from motion-captured data. The kinematics and EMG activity were analyzed for each stride based on the detected events. Main results Significant decreases in stride duration, stance and swing times and an increase in stride length were observed with increasing speed. A transition in gait pattern occurred at the 1.0m/s walking speed. Significant increases in the range of motion of the knee and ankle joints were observed at higher speeds. Also, the points of minimum and maximum joint angles occurred earlier in the gait cycle as the walking speed increased. The onset of EMG activity in the biceps femoris muscle occurred significantly earlier in the gait cycle with increasing speed. Significance YMPs are becoming frequently used as large animal models for preclinical testing and translation of novel interventions to humans. A comprehensive characterization of overground walking in neurologically-intact YMPs is provided in this study. These normative measures set the basis against which the effects of future interventions on locomotor capacity in YMPs can be compared.
Collapse
Affiliation(s)
- Soroush Mirkiani
- Neuroscience & Mental Health Institute and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, University of Alberta, Edmonton, Alberta, T6G 2R3, CANADA
| | - David A Roszko
- Neuroscience & Mental Health Institute and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Carly O'Sullivan
- Neuroscience & Mental Health Institute and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz, Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Pouria Faridi
- Neuroscience & Mental Health Institute and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - David S Hu
- Department of Medicine and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Daniel Fang
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Dirk G Everaert
- Department of Medicine and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Amirali Toossi
- Neuroscience & Mental Health Institute and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, Edmonton, Alberta, T6G 2R3, CANADA
| | - Peter E Konrad
- Department of Neurosurgery, West Virginia University, PO Box 9183, Morgantown, West Virginia, 26506, UNITED STATES
| | - Kevin Robinson
- School of Physical Therapy, Belmont University, 341 McWhorter Hall, Nashville, Tennessee, 37212, UNITED STATES
| | - Vivian K Mushahwar
- Department of Medicine and Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, 5005 Katz Building, University of Alberta, Edmonton, Alberta, T6G 2R3, CANADA
| |
Collapse
|
33
|
Mckean NE, Handley RR, Snell RG. A Review of the Current Mammalian Models of Alzheimer's Disease and Challenges That Need to Be Overcome. Int J Mol Sci 2021; 22:13168. [PMID: 34884970 PMCID: PMC8658123 DOI: 10.3390/ijms222313168] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is one of the looming health crises of the near future. Increasing lifespans and better medical treatment for other conditions mean that the prevalence of this disease is expected to triple by 2050. The impact of AD includes both the large toll on individuals and their families as well as a large financial cost to society. So far, we have no way to prevent, slow, or cure the disease. Current medications can only alleviate some of the symptoms temporarily. Many animal models of AD have been created, with the first transgenic mouse model in 1995. Mouse models have been beset by challenges, and no mouse model fully captures the symptomatology of AD without multiple genetic mutations and/or transgenes, some of which have never been implicated in human AD. Over 25 years later, many mouse models have been given an AD-like disease and then 'cured' in the lab, only for the treatments to fail in clinical trials. This review argues that small animal models are insufficient for modelling complex disorders such as AD. In order to find effective treatments for AD, we need to create large animal models with brains and lifespan that are closer to humans, and underlying genetics that already predispose them to AD-like phenotypes.
Collapse
Affiliation(s)
- Natasha Elizabeth Mckean
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Renee Robyn Handley
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Russell Grant Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
34
|
Schachtschneider KM, Schook LB, Meudt JJ, Shanmuganayagam D, Zoller JA, Haghani A, Li CZ, Zhang J, Yang A, Raj K, Horvath S. Epigenetic clock and DNA methylation analysis of porcine models of aging and obesity. GeroScience 2021; 43:2467-2483. [PMID: 34523051 PMCID: PMC8599541 DOI: 10.1007/s11357-021-00439-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022] Open
Abstract
DNA-methylation profiles have been used successfully to develop highly accurate biomarkers of age, epigenetic clocks, for many species. Using a custom methylation array, we generated DNA methylation data from n = 238 porcine tissues including blood, bladder, frontal cortex, kidney, liver, and lung, from domestic pigs (Sus scrofa domesticus) and minipigs (Wisconsin Miniature Swine™). Samples used in this study originated from Large White X Landrace crossbred pigs, Large White X Minnesota minipig crossbred pigs, and Wisconsin Miniature Swine™. We present 4 epigenetic clocks for pigs that are distinguished by their compatibility with tissue type (pan-tissue and blood clock) and species (pig and human). Two dual-species human-pig pan-tissue clocks accurately measure chronological age and relative age, respectively. We also characterized CpGs that differ between minipigs and domestic pigs. Strikingly, several genes implicated by our epigenetic studies of minipig status overlap with genes (ADCY3, TFAP2B, SKOR1, and GPR61) implicated by genetic studies of body mass index in humans. In addition, CpGs with different levels of methylation between the two pig breeds were identified proximal to genes involved in blood LDL levels and cholesterol synthesis, of particular interest given the minipig's increased susceptibility to cardiovascular disease compared to domestic pigs. Thus, breed-specific differences of domestic and minipigs may potentially help to identify biological mechanisms underlying weight gain and aging-associated diseases. Our porcine clocks are expected to be useful for elucidating the role of epigenetics in aging and obesity, and the testing of anti-aging interventions.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois At Chicago, Chicago, IL USA
- Department of Biochemistry and Molecular Genetics, University of Illinois At Chicago, Chicago, IL USA
- National Center for Supercomputing Applications, University of Illinois At Urbana-Champaign, Urban, IL USA
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois At Chicago, Chicago, IL USA
- Department of Animal Sciences, University of Illinois At Urbana-Champaign, Urbana, IL USA
| | - Jennifer J. Meudt
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin – Madison, Madison, WI USA
| | - Dhanansayan Shanmuganayagam
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin – Madison, Madison, WI USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Joseph A. Zoller
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Amin Haghani
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Caesar Z. Li
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Joshua Zhang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Gonda Building, 695 Charles Young Drive South, Los Angeles, CA 90095 USA
| | - Andrew Yang
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
| | - Ken Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, UK
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Gonda Building, 695 Charles Young Drive South, Los Angeles, CA 90095 USA
| |
Collapse
|
35
|
Strawn M, Moraes JGN, Safranski TJ, Behura SK. Sexually Dimorphic Transcriptomic Changes of Developing Fetal Brain Reveal Signaling Pathways and Marker Genes of Brain Cells in Domestic Pigs. Cells 2021; 10:2439. [PMID: 34572090 PMCID: PMC8466205 DOI: 10.3390/cells10092439] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/13/2022] Open
Abstract
In this study, transcriptomic changes of the developing brain of pig fetuses of both sexes were investigated on gestation days (GD) 45, 60 and 90. Pig fetal brain grows rapidly around GD60. Consequently, gene expression of the fetal brain was distinctly different on GD90 compared to that of GD45 and GD60. In addition, varying numbers of differentially expressed genes (DEGs) were identified in the male brain compared to the female brain during development. The sex of adjacent fetuses also influenced gene expression of the fetal brain. Extensive changes in gene expression at the exon-level were observed during brain development. Pathway enrichment analysis showed that the ionotropic glutamate receptor pathway and p53 pathway were enriched in the female brain, whereas specific receptor-mediated signaling pathways were enriched in the male brain. Marker genes of neurons and astrocytes were significantly differentially expressed between male and female brains during development. Furthermore, comparative analysis of gene expression patterns between fetal brain and placenta suggested that genes related to ion transportation may play a key role in the regulation of the brain-placental axis in pig. Collectively, the study suggests potential application of pig models to better understand influence of fetal sex on brain development.
Collapse
Affiliation(s)
- Monica Strawn
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (M.S.); (T.J.S.); (J.G.N.M.)
| | - Joao G. N. Moraes
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (M.S.); (T.J.S.); (J.G.N.M.)
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74075, USA
| | - Timothy J. Safranski
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (M.S.); (T.J.S.); (J.G.N.M.)
| | - Susanta K. Behura
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (M.S.); (T.J.S.); (J.G.N.M.)
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
36
|
Jarvis S, Koumadoraki E, Madouros N, Sharif S, Saleem A, Khan S. Non-rodent animal models of osteosarcoma: A review. Cancer Treat Res Commun 2021; 27:100307. [PMID: 33453605 DOI: 10.1016/j.ctarc.2021.100307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 11/29/2022]
Abstract
Osteosarcoma is extremely malignant, and the most common cancer that affects bone. Current treatments involve surgical resection of the affected area and multi-agent chemotherapy, though survival rate is generally poor for those affected by metastases. As treatment for osteosarcoma has remained unchanged for the past few decades, there is a need for further advancements in the understanding of osteosarcoma biology and therapeutics. Thus, reliable animal models that can accurately recapitulate the disease are required. Though rodents represent the most popular animal model of osteosarcoma, they may not model the disease best. This review analyzes emerging alternative non-rodent animal models of osteosarcoma, such as the chick chorioallantoic membrane (CAM) assay, pigs, and canines. Each of these alternatives offer advantages over classic rodent models for pre-clinical research. Research of these cross-species platforms imparts knowledge of metastases biology and potential new treatments for osteosarcoma.
Collapse
Affiliation(s)
- Sommer Jarvis
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States.
| | - Evgenia Koumadoraki
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States
| | - Nikolaos Madouros
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States
| | - Shayka Sharif
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States
| | - Amber Saleem
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States
| | - Safeera Khan
- California Institute of Neurosciences & Behavioral Psychology, 4751 Mangels Blvd, Fairfield, CA 94534, United States
| |
Collapse
|
37
|
Żakowski W. Animal Use in Neurobiological Research. Neuroscience 2020; 433:1-10. [PMID: 32156550 DOI: 10.1016/j.neuroscience.2020.02.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022]
Abstract
The fact that neurobiological research is reliant upon laboratory-reared rodents is well known. The following paper discusses this topic broadly, but also aims to highlight other species used in the study of the nervous system and the evolution of animal species usage from the end of World War II through recent investigations. Attention is drawn to the dramatic reduction in the diversity of species used in neuroscience, with a significant shift toward two species, the mouse (Mus musculus) and rat (Rattus norvegicus). Such a limitation in animal species causes many difficulties in the development of new therapies for various neuropsychiatric diseases. Based on numerous scientific publications, the advantages of using a greater diversity of species in neuroscience and the disadvantages of focusing on mice and rats are presented.
Collapse
Affiliation(s)
- Witold Żakowski
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| |
Collapse
|
38
|
Kalla D, Kind A, Schnieke A. Genetically Engineered Pigs to Study Cancer. Int J Mol Sci 2020; 21:E488. [PMID: 31940967 PMCID: PMC7013672 DOI: 10.3390/ijms21020488] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent decades have seen groundbreaking advances in cancer research. Genetically engineered animal models, mainly in mice, have contributed to a better understanding of the underlying mechanisms involved in cancer. However, mice are not ideal for translating basic research into studies closer to the clinic. There is a need for complementary information provided by non-rodent species. Pigs are well suited for translational biomedical research as they share many similarities with humans such as body and organ size, aspects of anatomy, physiology and pathophysiology and can provide valuable means of developing and testing novel diagnostic and therapeutic procedures. Porcine oncology is a new field, but it is clear that replication of key oncogenic mutation in pigs can usefully mimic several human cancers. This review briefly outlines the technology used to generate genetically modified pigs, provides an overview of existing cancer models, their applications and how the field may develop in the near future.
Collapse
Affiliation(s)
| | | | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany; (D.K.); (A.K.)
| |
Collapse
|