Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (290)

Search Parameters:
Keywords = genetically modified animal

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 690 KiB  
Review
Diabetes and Sarcopenia: Metabolomic Signature of Pathogenic Pathways and Targeted Therapies
by Anamaria Andreea Danciu, Cornelia Bala, Georgeta Inceu, Camelia Larisa Vonica, Adriana Rusu, Gabriela Roman and Dana Mihaela Ciobanu
Int. J. Mol. Sci. 2025, 26(15), 7574; https://doi.org/10.3390/ijms26157574 - 5 Aug 2025
Abstract
Diabetes mellites (DM) is a chronic disease with increasing prevalence worldwide and multiple health implications. Among them, sarcopenia is a metabolic disorder characterized by loss of muscle mass and function. The two age-related diseases, DM and sarcopenia, share underlying pathophysiological pathways. This narrative [...] Read more.
Diabetes mellites (DM) is a chronic disease with increasing prevalence worldwide and multiple health implications. Among them, sarcopenia is a metabolic disorder characterized by loss of muscle mass and function. The two age-related diseases, DM and sarcopenia, share underlying pathophysiological pathways. This narrative literature review aims to provide an overview of the existing evidence on metabolomic studies evaluating DM associated with sarcopenia. Advancements in targeted and untargeted metabolomics techniques could provide better insight into the pathogenesis of sarcopenia in DM and describe their entangled and fluctuating interrelationship. Recent evidence showed that sarcopenia in DM induced significant changes in protein, lipid, carbohydrate, and in energy metabolisms in humans, animal models of DM, and cell cultures. Newer metabolites were reported, known metabolites were also found significantly modified, while few amino acids and lipids displayed a dual behavior. In addition, several therapeutic approaches proved to be promising interventions for slowing the progression of sarcopenia in DM, including physical activity, newer antihyperglycemic classes, D-pinitol, and genetic USP21 ablation, although none of them were yet validated for clinical use. Conversely, ceramides had a negative impact. Further research is needed to confirm the utility of these findings and to provide potential metabolomic biomarkers that might be relevant for the pathogenesis and treatment of sarcopenia in DM. Full article
Show Figures

Figure 1

22 pages, 1078 KiB  
Review
The Cannabinoid Pharmacology of Bone Healing: Developments in Fusion Medicine
by Gabriel Urreola, Michael Le, Alan Harris, Jose A. Castillo, Augustine M. Saiz, Hania Shahzad, Allan R. Martin, Kee D. Kim, Safdar Khan and Richard Price
Biomedicines 2025, 13(8), 1891; https://doi.org/10.3390/biomedicines13081891 - 3 Aug 2025
Viewed by 330
Abstract
Background/Objectives: Cannabinoid use is rising among patients undergoing spinal fusion, yet its influence on bone healing is poorly defined. The endocannabinoid system (ECS)—through cannabinoid receptors 1 (CB1) and 2 (CB2)—modulates skeletal metabolism. We reviewed preclinical, mechanistic and clinical evidence to clarify how individual [...] Read more.
Background/Objectives: Cannabinoid use is rising among patients undergoing spinal fusion, yet its influence on bone healing is poorly defined. The endocannabinoid system (ECS)—through cannabinoid receptors 1 (CB1) and 2 (CB2)—modulates skeletal metabolism. We reviewed preclinical, mechanistic and clinical evidence to clarify how individual cannabinoids affect fracture repair and spinal arthrodesis. Methods: PubMed, Web of Science and Scopus were searched from inception to 31 May 2025 with the terms “cannabinoid”, “CB1”, “CB2”, “spinal fusion”, “fracture”, “osteoblast” and “osteoclast”. Animal studies, in vitro experiments and clinical reports that reported bone outcomes were eligible. Results: CB2 signaling was uniformly osteogenic. CB2-knockout mice developed high-turnover osteoporosis, whereas CB2 agonists (HU-308, JWH-133, HU-433, JWH-015) restored trabecular volume, enhanced osteoblast activity and strengthened fracture callus. Cannabidiol (CBD), a non-psychoactive phytocannabinoid with CB2 bias, accelerated early posterolateral fusion in rats and reduced the RANKL/OPG ratio without compromising final union. In contrast, sustained or high-dose Δ9-tetrahydrocannabinol (THC) activation of CB1 slowed chondrocyte hypertrophy, decreased mesenchymal-stromal-cell mineralization and correlated clinically with 6–10% lower bone-mineral density and a 1.8–3.6-fold higher pseudarthrosis or revision risk. Short-course or low-dose THC appeared skeletal neutral. Responses varied with sex, age and genetic background; no prospective trials defined safe perioperative dosing thresholds. Conclusions: CB2 activation and CBD consistently favor bone repair, whereas chronic high-THC exposure poses a modifiable risk for nonunion in spine surgery. Prospective, receptor-specific trials stratified by THC/CBD ratio, patient sex and ECS genotype are needed to establish evidence-based cannabinoid use in spinal fusion. Full article
(This article belongs to the Topic Cannabis, Cannabinoids and Its Derivatives)
Show Figures

Figure 1

39 pages, 675 KiB  
Review
Unlocking Implantation: The Role of Nitric Oxide, NO2-NO3, and eNOS in Endometrial Receptivity and IVF Success—A Systematic Review
by Charalampos Voros, Iwakeim Sapantzoglou, Despoina Mavrogianni, Diamantis Athanasiou, Antonia Varthaliti, Kyriakos Bananis, Antonia Athanasiou, Aikaterini Athanasiou, Anthi-Maria Papahliou, Constantinos G. Zografos, Athanasios Gkirgkinoudis, Ioannis Papapanagiotou, Kyriaki Migklis, Dimitris Mazis Kourakos, Georgios Papadimas, Maria Anastasia Daskalaki, Panagiotis Antsaklis, Dimitrios Loutradis and Georgios Daskalakis
Int. J. Mol. Sci. 2025, 26(14), 6569; https://doi.org/10.3390/ijms26146569 - 8 Jul 2025
Viewed by 524
Abstract
Nitric oxide (NO) predominantly regulates endometrial receptivity, angiogenesis, immunological tolerance, and trophoblast invasion throughout the implantation period. Both insufficient and excessive nitric oxide production have been linked to suboptimal embryo implantation and infertility. The primary enzymatic source of uterine nitric oxide, along with hormonal, [...] Read more.
Nitric oxide (NO) predominantly regulates endometrial receptivity, angiogenesis, immunological tolerance, and trophoblast invasion throughout the implantation period. Both insufficient and excessive nitric oxide production have been linked to suboptimal embryo implantation and infertility. The primary enzymatic source of uterine nitric oxide, along with hormonal, metabolic, and immunological variables and genetic variations in the endothelial nitric oxide synthase gene (NOS3), affects endothelial nitric oxide synthase (eNOS). Despite its considerable importance, there is limited knowledge regarding the practical implementation of nitric oxide-related diagnoses and therapies in reproductive medicine. A comprehensive assessment was performed in accordance with the PRISMA principles. Electronic searches were carried out in PubMed, Scopus, and Embase, and we analyzed the literature published from 2000 to 2024 regarding the association between NO, its metabolites (NO2 and NO3), eNOS expression, NOS3 gene variants, and reproductive outcomes. Relevant studies encompassed clinical trials, observational studies, and experimental research using either human or animal subjects. We collected data about therapeutic interventions, hormonal and immunological associations, nitric oxide measurement techniques, and in vitro fertilization success rates. A total of thirty-four studies were included. Dysregulated nitric oxide signaling, characterized by modified eNOS expression, oxidative stress, or NOS3 polymorphisms (e.g., Glu298Asp and intron 4 VNTR), was linked to diminished endometrial receptivity and an elevated risk of implantation failure and miscarriage. The dynamics of local uterine NO are essential as elevated and diminished systemic levels of NO2/NO3 corresponded with enhanced and decreased implantation rates, respectively. Among many therapeutic approaches, targeted hormone treatments, antioxidant therapy, and dietary nitrate supplements have demonstrated potential in restoring nitric oxide balance and enhancing reproductive outcomes. In animal models, the modification of nitric oxide significantly impacted decidualization, angiogenesis, and embryo viability. Nitric oxide is a multifaceted molecular mediator with considerable ramifications for successful implantation. Its therapeutic and diagnostic efficacy increases with its sensitivity to environmental, hormonal, and genetic alterations. Integrating targeted nitric oxide modulation, oxidative stress assessment, and NOS3 genotyping with personalized reproductive therapy will enhance endometrial receptivity and improve IVF outcomes. Future translational research should incorporate nitric oxide signaling into personalized treatment protocols for patients with unexplained infertility or recurrent implantation failure. Full article
(This article belongs to the Special Issue Molecular Advances in Obstetrical and Gynaecological Disorders)
Show Figures

Figure 1

23 pages, 331 KiB  
Review
Reviving the Dire Wolf? A Case Study in Welfare Ethics, Legal Gaps, and Ontological Ambiguity
by Alexandre Azevedo and Manuel Magalhães-Sant’Ana
Animals 2025, 15(13), 1839; https://doi.org/10.3390/ani15131839 - 21 Jun 2025
Viewed by 1072
Abstract
The recent birth of genetically modified canids phenotypically resembling the extinct dire wolf (Aenocyon dirus) was hailed as a landmark in synthetic biology. Using genome editing and cloning, the biotech company Colossal Biosciences created three such animals from gray wolf cells, [...] Read more.
The recent birth of genetically modified canids phenotypically resembling the extinct dire wolf (Aenocyon dirus) was hailed as a landmark in synthetic biology. Using genome editing and cloning, the biotech company Colossal Biosciences created three such animals from gray wolf cells, describing the project as an effort in “functional de-extinction”. This case raises significant questions regarding animal welfare, moral justification, and regulatory governance. We used the five domains model framework to assess the welfare risks for the engineered animals, the surrogate mothers used in reproduction, and other animals potentially affected by future reintroduction or escape scenarios. Ethical implications are examined through utilitarian, deontological, virtue, relational, and environmental ethics. Our analysis suggests that the project suffers from ontological ambiguity: it is unclear whether the animals created are resurrected species, hybrids, or novel organisms. While the current welfare of the engineered animals may be manageable, their long-term well-being, particularly under rewilding scenarios, is likely to be compromised. The moral arguments for reviving long-extinct species are weak, particularly in cases where extinction was not anthropogenic. Legally, the current EU frameworks lack the clarity and scope to classify, regulate, or protect genetically engineered extinct animals. We recommend that functional de-extinction involving sentient beings be approached with caution, supported by revised welfare tools and regulatory mechanisms. Full article
(This article belongs to the Special Issue Wild Animal Welfare: Science, Ethics and Law)
43 pages, 2656 KiB  
Review
α-Synuclein Pathology in Synucleinopathies: Mechanisms, Biomarkers, and Therapeutic Challenges
by Oscar Arias-Carrión, Magdalena Guerra-Crespo, Francisco J. Padilla-Godínez, Luis O. Soto-Rojas and Elías Manjarrez
Int. J. Mol. Sci. 2025, 26(11), 5405; https://doi.org/10.3390/ijms26115405 - 4 Jun 2025
Viewed by 1793
Abstract
Parkinson’s disease and related synucleinopathies, including dementia with Lewy bodies and multiple system atrophy, are characterised by the pathological aggregation of the α-synuclein (aSyn) protein in neuronal and glial cells, leading to cellular dysfunction and neurodegeneration. This review synthesizes knowledge of aSyn biology, [...] Read more.
Parkinson’s disease and related synucleinopathies, including dementia with Lewy bodies and multiple system atrophy, are characterised by the pathological aggregation of the α-synuclein (aSyn) protein in neuronal and glial cells, leading to cellular dysfunction and neurodegeneration. This review synthesizes knowledge of aSyn biology, including its structure, aggregation mechanisms, cellular interactions, and systemic influences. We highlight the structural diversity of aSyn aggregates, ranging from oligomers to fibrils, their strain-like properties, and their prion-like propagation. While the role of prion-like mechanisms in disease progression remains a topic of ongoing debate, these processes may contribute to the clinical heterogeneity of synucleinopathies. Dysregulation of protein clearance pathways, including chaperone-mediated autophagy and the ubiquitin–proteasome system, exacerbates aSyn accumulation, while post-translational modifications influence its toxicity and aggregation propensity. Emerging evidence suggests that immune responses and alterations in the gut microbiome are key modulators of aSyn pathology, linking peripheral processes—particularly those of intestinal origin—to central neurodegeneration. Advances in biomarker development, such as cerebrospinal fluid assays, post-translationally modified aSyn, and real-time quaking-induced conversion technology, hold promise for early diagnosis and disease monitoring. Furthermore, positron emission tomography imaging and conformation-specific antibodies offer innovative tools for visualising and targeting aSyn pathology in vivo. Despite significant progress, challenges remain in accurately modelling human synucleinopathies, as existing animal and cellular models capture only specific aspects of the disease. This review underscores the need for more reliable aSyn biomarkers to facilitate the development of effective treatments. Achieving this goal requires an interdisciplinary approach integrating genetic, epigenetic, and environmental insights. Full article
(This article belongs to the Special Issue Molecular Insights in Neurodegeneration)
Show Figures

Graphical abstract

31 pages, 1374 KiB  
Review
Genetically Modified Animal-Derived Products: From Regulations to Applications
by Carlos Fajardo, Marta Macedo, Tonka Buha, Marcos De Donato, Benjamin Costas and Juan Miguel Mancera
Animals 2025, 15(11), 1570; https://doi.org/10.3390/ani15111570 - 27 May 2025
Viewed by 1108
Abstract
Biotechnological advances applied to the generation of genetically modified (GM) animals have shown the potential to develop innovative solutions for different challenges in key areas such as agriculture and human medicine. Despite its enormous potential, the deployment of genetic modification in animals, and [...] Read more.
Biotechnological advances applied to the generation of genetically modified (GM) animals have shown the potential to develop innovative solutions for different challenges in key areas such as agriculture and human medicine. Despite its enormous potential, the deployment of genetic modification in animals, and its subsequent commercialization, does not meet the same public acceptance as GM plant-derived products, which are currently widely adopted around the world. In this review, we highlight the main examples of GM and gene-edited animal-derived products already approved by the FDA and discuss the regulatory context inherent to such processes, including the risk-based assessment analysis based on a case-by-case evaluation. Moreover, cases of GM animals already approved by other jurisdictions around the world are also discussed. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

16 pages, 4131 KiB  
Article
Identification, Pathogenicity, and Reverse Genetics System Construction of a Pseudorabies Virus Isolate from Pigs in China
by Mo Zhou, Haiyang Liang, Nannan Nie, Li Zhang, Rui Zhu, Shinuo Cao and Shanyuan Zhu
Vet. Sci. 2025, 12(6), 519; https://doi.org/10.3390/vetsci12060519 - 26 May 2025
Viewed by 530
Abstract
Pseudorabies virus (PRV) is a highly contagious pathogen in swine that can cross species barriers and infect other mammals, including humans. Given the potential for interspecies transmission and its threat to public health, understanding the molecular biology of PRV strains is essential for [...] Read more.
Pseudorabies virus (PRV) is a highly contagious pathogen in swine that can cross species barriers and infect other mammals, including humans. Given the potential for interspecies transmission and its threat to public health, understanding the molecular biology of PRV strains is essential for developing effective control measures and preparing for future pandemics. In this study, a novel PRV strain, PRV-HL-2021, was isolated from an outbreak in Heilongjiang Province, China. The viral genome was used to establish a reverse genetics system based on a fosmid library of the PRV-HL-2021 genome. This system facilitated the creation of recombinant PRV, including one expressing EGFP and another with deletions in the US9, gI, and gE genes. PRV-HL-2021 was found to be highly lethal to mice in vivo. The recombinant PRV strains, such as rPRV-US9-EGFP and rPRV-delgI/gE/US9, exhibited growth characteristics similar to the parental PRV-HL-2021 strain. The isolation and characterization of PRV-HL-2021 contribute to a better understanding of the genetic diversity of PRV strains. The developed reverse genetics system provides valuable tools for investigating viral functions, creating genetically modified PRV strains, and advancing the development of safer vaccines. These findings will enhance strategies for controlling PRV outbreaks and mitigating its impact on both animal and public health. Full article
Show Figures

Figure 1

53 pages, 1175 KiB  
Review
Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy
by Pierre Bougnères and Catherine Le Stunff
Genes 2025, 16(5), 590; https://doi.org/10.3390/genes16050590 - 17 May 2025
Cited by 2 | Viewed by 1570
Abstract
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal [...] Read more.
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. Methods: This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. Results: In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. Conclusions: Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

41 pages, 2878 KiB  
Review
Modeling Alzheimer’s Disease: A Review of Gene-Modified and Induced Animal Models, Complex Cell Culture Models, and Computational Modeling
by Anna M. Timofeeva, Kseniya S. Aulova and Georgy A. Nevinsky
Brain Sci. 2025, 15(5), 486; https://doi.org/10.3390/brainsci15050486 - 5 May 2025
Viewed by 1879
Abstract
Alzheimer’s disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer’s disease and facilitate therapeutic development. Mammals, [...] Read more.
Alzheimer’s disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer’s disease and facilitate therapeutic development. Mammals, especially mice, are the most common models for studying the pathogenesis of this disease in vivo. To date, the scientific literature has documented more than 280 mouse models exhibiting diverse aspects of Alzheimer’s disease pathogenesis. Other mammalian species, including rats, pigs, and primates, have also been utilized as models. Selected aspects of Alzheimer’s disease have also been modeled in simpler model organisms, such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio. It is possible to model Alzheimer’s disease not only by creating genetically modified animal lines but also by inducing symptoms of this neurodegenerative disease. This review discusses the main methods of creating induced models, with a particular focus on modeling Alzheimer’s disease on cell cultures. Induced pluripotent stem cell (iPSC) technology has facilitated novel investigations into the mechanistic underpinnings of diverse diseases, including Alzheimer’s. Progress in culturing brain tissue allows for more personalized studies on how drugs affect the brain. Recent years have witnessed substantial advancements in intricate cellular system development, including spheroids, three-dimensional scaffolds, and microfluidic cultures. Microfluidic technologies have emerged as cutting-edge tools for studying intercellular interactions, the tissue microenvironment, and the role of the blood–brain barrier (BBB). Modern biology is experiencing a significant paradigm shift towards utilizing big data and omics technologies. Computational modeling represents a powerful methodology for researching a wide array of human diseases, including Alzheimer’s. Bioinformatic methodologies facilitate the analysis of extensive datasets generated via high-throughput experimentation. It is imperative to underscore the significance of integrating diverse modeling techniques in elucidating pathogenic mechanisms in their entirety. Full article
Show Figures

Figure 1

19 pages, 1261 KiB  
Review
CRISPR-Cas9 in the Tailoring of Genetically Engineered Animals
by Wiktoria Urban, Marta Kropacz, Maksymilian Łach and Anna Jankowska
Curr. Issues Mol. Biol. 2025, 47(5), 330; https://doi.org/10.3390/cimb47050330 - 4 May 2025
Cited by 1 | Viewed by 2780
Abstract
CRISPR-Cas9 enables targeted genome editing and has become a pivotal tool in biomedical research and animal genome engineering. This review highlights its application in generating genetically modified animals used as preclinical disease models, bioreactors for recombinant protein production, and potential sources of xenotransplantation [...] Read more.
CRISPR-Cas9 enables targeted genome editing and has become a pivotal tool in biomedical research and animal genome engineering. This review highlights its application in generating genetically modified animals used as preclinical disease models, bioreactors for recombinant protein production, and potential sources of xenotransplantation organs. We also discuss its role in improving livestock traits, welfare, and breeding efficiency. The benefits and limitations of CRISPR-Cas9 are examined, emphasizing its transformative potential in research and agricultural biotechnology. Full article
(This article belongs to the Special Issue Technological Advances Around Next-Generation Sequencing Application)
Show Figures

Figure 1

24 pages, 4838 KiB  
Article
Genetically Modified Mesenchymal Stromal/Stem Cells as a Delivery Platform for SE-33, a Cathelicidin LL-37 Analogue: Preclinical Pharmacokinetics and Tissue Distribution in C57BL/6 Mice
by Vagif Ali oglu Gasanov, Dmitry Alexandrovich Kashirskikh, Victoria Alexandrovna Khotina, Arthur Anatolievich Lee, Sofya Yurievna Nikitochkina, Daria Mikhailovna Kuzmina, Irina Vasilievna Mukhina, Ekaterina Andreevna Vorotelyak and Andrey Valentinovich Vasiliev
Antibiotics 2025, 14(5), 429; https://doi.org/10.3390/antibiotics14050429 - 24 Apr 2025
Viewed by 601
Abstract
Background: The genetic modification of mesenchymal stromal/stem cells (MSCs) to express antimicrobial peptides may provide a promising strategy for developing advanced cell-based therapies for bacterial infections, including those caused or complicated by antibiotic-resistant bacteria. We have previously demonstrated that genetically modified Wharton’s jelly-derived [...] Read more.
Background: The genetic modification of mesenchymal stromal/stem cells (MSCs) to express antimicrobial peptides may provide a promising strategy for developing advanced cell-based therapies for bacterial infections, including those caused or complicated by antibiotic-resistant bacteria. We have previously demonstrated that genetically modified Wharton’s jelly-derived MSCs expressing an antimicrobial peptide SE-33 (WJ-MSC-SE33) effectively reduce bacterial load, inflammation, and mortality in a mouse model of Staphylococcus aureus-induced pneumonia compared with native WJ-MSCs. The present study aimed to evaluate the pharmacokinetics and tissue distribution of the SE-33 peptide expressed by WJ-MSC-SE33 following administration to animals. Methods: WJ-MSC-SE33 were administered to C57BL/6 mice at therapeutic and excess doses. The biodistribution and pharmacokinetics of the SE-33 peptide were analyzed in serum, lungs, liver, and spleen using chromatographic methods after single and repeated administrations. Results: The SE-33 peptide exhibited dose-dependent pharmacokinetics. The highest levels of SE-33 peptide were detected in the liver and lungs, with persistence in tissues for up to 48 h at medium and high doses of administered WJ-MSC-SE33. A repeated administration of WJ-MSC-SE33 increased SE-33 levels in target organs. Conclusions: The SE-33 peptide expressed by genetically modified WJ-MSCs demonstrated predictable pharmacokinetics and effective biodistribution. These findings, together with the previously established safety profile of WJ-MSC-SE33, support its potential as a promising cell-based therapy for bacterial infections, particularly those associated with antibiotic resistance. Full article
Show Figures

Figure 1

11 pages, 892 KiB  
Article
Visualization of Runs of Homozygosity and Classification Using Convolutional Neural Networks
by Siroj Bakoev, Maria Kolosova, Timofey Romanets, Faridun Bakoev, Anatoly Kolosov, Elena Romanets, Anna Korobeinikova, Ilona Bakoeva, Vagif Akhmedli and Lyubov Getmantseva
Biology 2025, 14(4), 426; https://doi.org/10.3390/biology14040426 - 16 Apr 2025
Viewed by 793
Abstract
Runs of homozygosity (ROH) are key elements of the genetic structure of populations, reflecting inbreeding levels, selection history, and potential associations with phenotypic traits. This study proposes a novel approach to ROH analysis through visualization and classification using convolutional neural networks (CNNs). Genetic [...] Read more.
Runs of homozygosity (ROH) are key elements of the genetic structure of populations, reflecting inbreeding levels, selection history, and potential associations with phenotypic traits. This study proposes a novel approach to ROH analysis through visualization and classification using convolutional neural networks (CNNs). Genetic data from Large White (n = 568) and Duroc (n = 600) pigs were used to construct ROH maps, where each homozygous segment was classified by length and visualized as a color-coded image. The analysis was conducted in two stages: (1) classification of animals by breed based on ROH maps and (2) identification of the presence or absence of a phenotypic trait (limb defects). Genotyping was performed using the GeneSeek® GGP SNP80x1_XT chip (Illumina Inc., San Diego, CA, USA), and ROH segments were identified using the software tool PLINK v1.9. To visualize individual maps, we utilized a modified function from the HandyCNV package. The results showed that the CNN model achieved 100% accuracy, sensitivity, and specificity in classifying pig breeds based on ROH maps. When analyzing the binary trait (presence or absence of limb defects), the model demonstrated an accuracy of 78.57%. Despite the moderate accuracy in predicting the phenotypic trait, the high negative predictive value (84.62%) indicates the model’s reliability in identifying healthy animals. This method can be applied not only in animal breeding research but also in medicine to study the association between ROH and hereditary diseases. Future plans include expanding the method to other types of genetic data and developing mechanisms to improve the interpretability of deep learning models. Full article
(This article belongs to the Special Issue Machine Learning Applications in Biology—2nd Edition)
Show Figures

Figure 1

37 pages, 21808 KiB  
Article
Preclinical Evaluation of the Safety, Toxicity and Efficacy of Genetically Modified Wharton’s Jelly Mesenchymal Stem/Stromal Cells Expressing the Antimicrobial Peptide SE-33
by Vagif Ali oglu Gasanov, Dmitry Alexandrovich Kashirskikh, Victoria Alexandrovna Khotina, Daria Mikhailovna Kuzmina, Sofya Yurievna Nikitochkina, Irina Vasilievna Mukhina, Ekaterina Andreevna Vorotelyak and Andrey Valentinovich Vasiliev
Cells 2025, 14(5), 341; https://doi.org/10.3390/cells14050341 - 26 Feb 2025
Cited by 1 | Viewed by 1015
Abstract
Mesenchymal stem/stromal cells (MSCs) offer promising therapeutic potential in cell-based therapies for various diseases. However, the safety of genetically modified MSCs remains poorly understood. This study aimed to evaluate the general toxicity and safety of Wharton’s Jelly-Derived MSCs (WJ-MSCs) engineered to express the [...] Read more.
Mesenchymal stem/stromal cells (MSCs) offer promising therapeutic potential in cell-based therapies for various diseases. However, the safety of genetically modified MSCs remains poorly understood. This study aimed to evaluate the general toxicity and safety of Wharton’s Jelly-Derived MSCs (WJ-MSCs) engineered to express the antimicrobial peptide SE-33 in an animal model. Genetically modified WJ-MSCs expressing SE-33 were administered to C57BL/6 mice at both therapeutic and excessive doses, either once or repeatedly. Animal monitoring included mortality, clinical signs, and behavioral observations. The toxicity assessment involved histopathological, hematological, and biochemical analyses of major organs and tissues, while immunotoxicity and immunogenicity were examined through humoral and cellular immune responses, macrophage phagocytic activity, and lymphocyte blast transformation. Antimicrobial efficacy was evaluated in a Staphylococcus aureus-induced pneumonia model by monitoring animal mortality and assessing bacterial load and inflammatory processes in the lungs. Mice receiving genetically modified WJ-MSCs exhibited no acute or chronic toxicity, behavioral abnormalities, or pathological changes, regardless of the dose or administration frequency. No significant immunotoxicity or alterations in immune responses were observed, and there were no notable changes in hematological or biochemical serum parameters. Infected animals treated with WJ-MSC-SE33 showed a significant reduction in bacterial load and lung inflammation and improved survival compared to control groups, demonstrating efficacy over native WJ-MSCs. Our findings suggest that WJ-MSCs expressing SE-33 are well tolerated, displaying a favorable safety profile comparable to native WJ-MSCs and potent antimicrobial activity, significantly reducing bacterial load, inflammation, and mortality in an S. aureus pneumonia model. These data support the safety profile of WJ-MSCs expressing SE-33 as a promising candidate for cell-based therapies for bacterial infections, particularly those complicated by antibiotic resistance. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Graphical abstract

20 pages, 3981 KiB  
Article
Field Investigation Evaluating the Efficacy of Porcine Reproductive and Respiratory Syndrome Virus Type 2 (PRRSV-2) Modified Live Vaccines in Nursery Pigs Exposed to Multiple Heterologous PRRSV Strains
by Sunit Mebumroong, Hongyao Lin, Patumporn Jermsutjarit, Angkana Tantituvanont and Dachrit Nilubol
Animals 2025, 15(3), 428; https://doi.org/10.3390/ani15030428 - 4 Feb 2025
Viewed by 2097
Abstract
This study was conducted to evaluate the protective efficacy of modified live vaccines (MLVs) against porcine reproductive and respiratory syndrome (PRRS) in nursery pigs in a worst case scenario where MLV does not match the genetic profile of the field isolate, different MLVs [...] Read more.
This study was conducted to evaluate the protective efficacy of modified live vaccines (MLVs) against porcine reproductive and respiratory syndrome (PRRS) in nursery pigs in a worst case scenario where MLV does not match the genetic profile of the field isolate, different MLVs are used for sows and piglets, and piglets are naturally exposed to genetically distinct heterologous PRRS virus (PRRSV) isolates. We divided 76,075, 2-week-old piglets from a seropositive sow herd vaccinated with US1-MLV into four groups. US1-MLV, US2-MLV, and US3-MLV groups were vaccinated with PRRSV-2 MLV including Ingelvac® PRRS MLV (Boehringer Ingelheim, Ingelheim am Rhein, Germany), HP-PRRSV-2 based MLV (Harbin Veterinary Research Institute, CAAS, Harbin, China), and Prime Pac® PRRS (MSD Animal Health, Rahway, NJ, USA), respectively. The NonVac group was left unvaccinated. At 0, 14, 28, and 56 days post-vaccination (DPV), sera were assayed for the presence of PRRSV-specific antibodies using ELISA and serum neutralization (SN), and PRRSV RNA using PCR. Average daily gain (ADG) and survival rates were compared between treatment groups. The results demonstrated vaccinated groups significantly improved in ADG compared to the non-vaccinated control group. Only US1-MLV and US3-MLV were able to significantly reduce mortality associated with field PRRSV infection in nursery pigs. Pigs vaccinated with US3-MLV displayed significantly lower mortality and higher ADG compared to all other groups. Field isolates were isolated and genetically compared to all three MLV vaccines at the start of the trial. The MLV with closest genetic similarity to the field isolate was US2-MLV by ORF5 gene comparison. This provided the lowest protection judging by ADG improvement and mortality reduction, as compared to US1-MLV and US3-MLV. Separately, strains of Thai PRRSV-2 isolates collected in 2017, 2019, and 2020 in the study area were investigated for evolutionary changes. Over time, we observed a shift in PRRSV-2 isolates from lineage 8.7 to lineage 1. The field isolates found shared 82.59–84.42%, 83.75–85.74%, and 84.25–85.90% nucleotide identity with the US1-MLV, US3-MLV and US2-MLV based vaccine, respectively. Our findings suggest genetic similarity between field viruses and vaccine strains should not be used as a predictor of field performance. We found that zootechnical performance of piglets was best in US3-MLV, despite sows being treated with a different vaccine The results also support that different MLVs can be used at different stages of production. Finally, we concluded that the shift from lineage 8.7 to lineage 1 was due to shifts in the worldwide prevalence of PRRSV isolates during that period of time and not due to vaccine recombination between isolates. Overall, MLV vaccine selection should be based on production performance and safety profile. Full article
(This article belongs to the Section Pigs)
Show Figures

Figure 1

20 pages, 2381 KiB  
Article
Reliable Polymerase Chain Reaction Methods for Screening for Porcine Endogenous Retroviruses-C (PERV-C) in Pigs
by Hina Jhelum, Dusan Kunec, Vasileios Papatsiros, Benedikt B. Kaufer and Joachim Denner
Viruses 2025, 17(2), 164; https://doi.org/10.3390/v17020164 - 24 Jan 2025
Viewed by 1018
Abstract
Porcine endogenous retrovirus C (PERV-C) is a gammaretrovirus present in the genome of many, but not all, pigs. It is an ecotropic virus, able to infect only pig cells. In contrast, PERV-A and PERV-B, which are present in all pigs, can infect cells [...] Read more.
Porcine endogenous retrovirus C (PERV-C) is a gammaretrovirus present in the genome of many, but not all, pigs. It is an ecotropic virus, able to infect only pig cells. In contrast, PERV-A and PERV-B, which are present in all pigs, can infect cells of multiple host species, including humans, thereby posing a risk for xenotransplantation when pigs are used as donor animals. Notably, PERV-C can recombine with PERV-A to produce PERV-A/C recombinants that can infect human cells and replicate to higher titers compared to the paternal PERV-A. The objective of this study is to evaluate the reliability of both existing and newly developed polymerase chain reactions (PCR) methods for detecting PERV-C, with the aim of selecting PERV-C-free pigs to be used for xenotransplantation. To detect PERV-C by PCR, specific primers targeting the region of the envelope protein gene, which differs from that of PERV-A and PERV-B due to its unique receptor binding site, must be employed. In this study, new PCR assays were developed to detect PERV-C and a total of ten PCR assays and one real-time PCR assay were evaluated for their reliability in detecting PERV-C. These assays were used to screen indigenous Greek black pigs, Auckland Island pigs, and German slaughterhouse pigs. Two of the PCR assays consistently yielded reliable results, whereas the other PCRs and the real-time PCR gave false positive results. Using the reliable assays, it was shown that one out of four indigenous Greek black pigs (using the same method in a previous publication 11 of 21 pigs were found PERV-C-negative), one out of ten German slaughterhouse pigs, the pig kidney cell line PK15, and all the Auckland Island pigs were PERV-C-negative. The reliable PCR assays will enable the screening of PERV-C-negative donor pigs to be used in xenotransplantation. Most importantly, all the Auckland Island pigs that were genetically modified in Germany for use in clinical trials were PERV-C-negative. Full article
(This article belongs to the Special Issue Porcine Viruses 2024)
Show Figures

Figure 1

Back to TopTop