1
|
Fernandes VS, Caballero R, Siguero-Álvarez M, Papoutsi T, Gimeno-Blanes JR, Delpón E, de la Pompa JL. Cardiac electrical abnormalities in a mouse model of left ventricular non-compaction cardiomyopathy. PLoS One 2025; 20:e0314840. [PMID: 40334239 PMCID: PMC12058163 DOI: 10.1371/journal.pone.0314840] [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] [Received: 11/22/2024] [Accepted: 03/05/2025] [Indexed: 05/09/2025] Open
Abstract
Mutations in MINDBOMB 1 (MIB1), encoding an E3 ubiquitin ligase of the NOTCH signaling pathway, cause left ventricular noncompaction cardiomyopathy (LVNC) in mice and humans, increasing the risk of arrhythmia and left ventricular dysfunction. This study aimed to investigate the effect of MIB1 mutations on cardiac electrical activity. We examined male Mib1flox;Tnnt2Cre mice, a disease model of LVNC, and wildtype littermates on the C57BL/6J genetic background. Our results demonstrate that the gap-junction protein connexin43 was delocalized from the intercalated disks to the lateral long axis of Mib1flox;Tnnt2Cre cardiomyocytes. Cardiomyocyte electrophysiology revealed an increase in the Na (INa) peak density at potentials between -50 and -30 mV in Mib1flox;Tnnt2Cre mice, with no changes in INa activation or inactivation kinetics. Mib1flox;Tnnt2Cre cardiomyocytes also showed decreases in outward K+ peak currents and currents at the end of depolarizing pulses at potentials ≥-10 mV and ≥-20 mV, respectively, and this was accompanied by a lower charge density at ≥-20 mV. Action potential duration was increased in Mib1flox;Tnnt2Cre cardiomyocytes. The cardiac stress, induced by swimming endurance training or β-adrenergic stimulation with isoproterenol, increases QTc duration in Mib1flox;Tnnt2Cre mice, accompanied by a decrease in T-wave amplitude and area. Swimming endurance training decreased heart rate in wildtype and Mib1flox;Tnnt2Cre mice but was unaffected by long-term isoproterenol treatment. These mouse findings are in agreement with an increased QTc duration found in LVNC patients carrying MIB1 mutations. These results provide insight into the outcomes of LVNC and relate its pathogenicity to impaired ventricular repolarization.
Collapse
Affiliation(s)
- Vítor S. Fernandes
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Caballero
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Marcos Siguero-Álvarez
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Tania Papoutsi
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan Ramón Gimeno-Blanes
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Unidad CSUR de Cardiopatías Familiares, Servicio de Cardiología, Hospital Universitario Virgen de la Arrixaca, Universidad de Murcia, Murcia, Spain
| | - Eva Delpón
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - José Luís de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Stephens VR, Ameli S, Major AS, Wanjalla CN. Mouse Models of HIV-Associated Atherosclerosis. Int J Mol Sci 2025; 26:3417. [PMID: 40244289 PMCID: PMC11989901 DOI: 10.3390/ijms26073417] [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: 03/04/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Several factors are implicated in the pathogenesis of CVD, and efforts have been made to reduce traditional risks, yet CVD remains a complex burden. Notably, people living with HIV (PLWH) are twice as likely to develop CVD compared to persons without HIV (PWoH). Intensive statin therapy, the first-line treatment to prevent cardiovascular events, is effective at reducing morbidity and mortality. However, statin therapy has not reduced the overall prevalence of CVD. Despite antiretroviral therapy (ART), and new guidelines for statin use, PLWH have persistent elevation of inflammatory markers, which is suggested to be a bigger driver of future cardiovascular events than low-density lipoprotein. Herein, we have summarized the development of atherosclerosis and highlighted mouse models of atherosclerosis in the presence and absence of HIV. Since most mouse strains have several mechanisms that are atheroprotective, researchers have developed mouse models to study CVD using dietary and genetic manipulations. In evaluating the current methodologies for studying HIV-associated atherosclerosis, we have detailed the benefits of integrating multi-omics analyses, genetic manipulations, and immune cell profiling within mouse models. These advanced approaches significantly enhance our capacity to address critical gaps in understanding the immune mechanisms driving CVD, including in the context of HIV.
Collapse
Affiliation(s)
- Victoria R. Stephens
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amy S. Major
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Tennessee Valley Health System, Department of Veterans Affairs, Nashville, TN 37212, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
3
|
Schmitz AM, Bumbaru SM, Fakhouri LS, Zhang DQ. Long-Term Impairment of Retinal Ganglion Cell Function After Oxygen-Induced Retinopathy. Cells 2025; 14:512. [PMID: 40214465 PMCID: PMC11988018 DOI: 10.3390/cells14070512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Premature infants with retinopathy of prematurity (ROP) have neovascularization of the retina, potentially resulting in low vision and even blindness. Some of these infants still have visual impairment, even if ROP resolves as they age. However, the mechanisms underlying the visual problems post-ROP are poorly understood. Because the pathological neovascularization in ROP infants can be mimicked in a mouse model with oxygen-induced retinopathy (OIR), we recapitulated post-ROP with post-OIR mice a few months after spontaneous regression of retinal neovascularization. Our pattern electroretinogram test demonstrates that post-OIR mice exhibit reduced P1-N2 responses, suggesting the impairment of retinal ganglion cells, the retina's output neurons. However, immunohistochemistry reveals that the density of retinal ganglion cells remains unchanged in post-OIR mice, indicating that the aforementioned pattern electroretinogram changes are functional. Our data further demonstrate that both light-adapted ex vivo electroretinogram a-waves (cone responses) and in vivo electroretinogram b-waves (ON cone bipolar cell responses) were significantly impaired in post-OIR mice. These results suggest that post-OIR impairment of the retinal cone pathway appears to result in the dysfunction of retinal ganglion cells, contributing to visual problems. A similar cellular mechanism could occur in post-ROP children, which is responsible for their visual impairment.
Collapse
Affiliation(s)
- Adam M. Schmitz
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA; (A.M.S.); (S.M.B.); (L.S.F.)
| | - Stephanie M. Bumbaru
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA; (A.M.S.); (S.M.B.); (L.S.F.)
| | - Laith S. Fakhouri
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA; (A.M.S.); (S.M.B.); (L.S.F.)
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA; (A.M.S.); (S.M.B.); (L.S.F.)
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
4
|
Giammona A, Terribile G, Rainone P, Pellizzer C, Porro D, Cerasa A, Sancini G, Rashid AU, Belloli S, Valtorta S, Lo Dico A, Bertoli G. Effects of particulate air pollution exposure on lung-brain axis and related miRNAs modulation in mouse models. Front Cell Dev Biol 2025; 13:1526424. [PMID: 40248351 PMCID: PMC12003928 DOI: 10.3389/fcell.2025.1526424] [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: 11/11/2024] [Accepted: 02/27/2025] [Indexed: 04/19/2025] Open
Abstract
Particulate matter exposure is linked to numerous health issues, including respiratory, cardiovascular, and neurodegenerative diseases. This review focuses on the biological mechanisms through which air pollution influences the lung-brain axis, highlighting the role of miRNAs in regulating gene pathways affected by PM. Some microRNAs (miRNAs) are identified as key modulators of cellular processes, including inflammation, epithelial-to-mesenchymal transition (EMT), and blood-brain barrier integrity. Using mice models to study these effects allows for controlled experimentation on the systemic distribution of PM across biological barriers. Among the imaging technologies, Positron Emission Tomography is the best approach to monitor the distribution and effects of PM in vivo. The research underscores the importance of miRNA profiles as potential markers for the health effects of PM exposure, suggesting that specific miRNAs could serve as early indicators of damage to the lung-brain axis.
Collapse
Affiliation(s)
- Alessandro Giammona
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Giulia Terribile
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Earth and Environmental Sciences, POLARIS Research Centre, University of Milano-Bicocca, Milano, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Paolo Rainone
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Chiara Pellizzer
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
| | - Danilo Porro
- PhD Program, Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milano, Italy
| | - Antonio Cerasa
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
| | - Giulio Sancini
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Earth and Environmental Sciences, POLARIS Research Centre, University of Milano-Bicocca, Milano, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Ameen-Ur Rashid
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
- PhD Program, Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milano, Italy
| | - Sara Belloli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Silvia Valtorta
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Alessia Lo Dico
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Gloria Bertoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| |
Collapse
|
5
|
Rajalekshmi R, Agrawal DK. Advancing Osteoarthritis Research: Insights from Rodent Models and Emerging Trends. JOURNAL OF ORTHOPAEDICS AND SPORTS MEDICINE 2025; 7:110-128. [PMID: 40264810 PMCID: PMC12014194 DOI: 10.26502/josm.511500187] [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: 04/24/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects millions of individuals worldwide, causing pain, disability, and a significant burden on public health. Preclinical research using animal models is essential to our understanding of the underlying pathogenesis of OA and developing therapeutic strategies. Rodent models, in particular, have become indispensable in studying OA due to their ability to mimic various features of human disease. This review provides an overview of commonly used rodent models of OA, including surgical induction (e.g., destabilization of the medial meniscus and anterior cruciate ligament transection), chemical induction (e.g., monoiodoacetate-induced cartilage degeneration), and genetically modified models. Additionally, age-related OA models that naturally develop OA-like symptoms in aged rodents are also discussed. Despite their utility, rodent models face limitations in fully recapitulating the complexity of human OA. Emerging trends in OA research, including the use of 3D imaging for joint analysis, molecular profiling for deeper insights into disease mechanisms, and advancements in biomarkers for early detection and treatment, are highlighted. These innovations provide new opportunities to refine existing models and enhance the translation of findings to clinical therapies. This critical review provides comprehensive information for researchers working in OA and related fields, promoting a better understanding of the available rodent models and their applications in OA research.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
6
|
Padash Barmchi M, Hassan RN, Afkhami M, Masly JP, Brown H, Collins QP, Grunsted MJ. Drosophila model of HPV18-Induced pathogenesis reveals a role for E6 oncogene in regulation of NF-κB and Wnt to inhibit apoptosis. Tumour Virus Res 2025; 19:200316. [PMID: 40074036 PMCID: PMC12008589 DOI: 10.1016/j.tvr.2025.200316] [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: 01/02/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025] Open
Abstract
Cancers caused by high-risk human papillomavirus (HPV) remain a significant health threat resulting in more than 300,000 deaths, annually. Persistent expression of two HPV oncogenes, E6 and E7, are necessary for cancer development and progression. E6 has several functions contributing to tumorigenesis one of which is blocking programmed cell death, apoptosis. The detailed mechanism of anti-apoptosis function of E6 is not fully understood. Here, using a Drosophila model of HPV18E6 and the human UBE3A-induced pathogenesis, we show that anti-apoptotic function of E6 is conserved in Drosophila. We demonstrate that the Drosophila homologs of human NF-κB transcription factors, Dorsal and Dif are proapoptotic. They induce the expression of Wingless (Wg, the Drosophila homolog of human Wnt), leading to apoptosis. Our results indicate that E6 oncogene inhibits apoptosis by downregulating the expression of Wg, Dorsal, and Dif. Additionally, we find that Dorsal and Dif, not only promote apoptosis but also regulate autophagy and necrosis. Dorsal promotes autophagy while Dif counteracts it, inducing the formation of acidic vacuoles and necrosis. Interestingly, although E6 blocks the proapoptotic function of Dorsal and Dif, it lacks the ability to interfere with their role in apoptosis-independent cell death. Given the high conservation of NF-κB transcription factors our results provide new insight into potential mechanisms mediated by NF-κB to intervene with cell immortalization action of E6 oncoprotein in HPV-infected cells.
Collapse
Affiliation(s)
| | - Rami N Hassan
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| | - Mehrnaz Afkhami
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - John P Masly
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| | - Harrison Brown
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Quincy P Collins
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, BC, Canada
| | - Michael J Grunsted
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
7
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2025; 36:262-277. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
8
|
Matera MG, Calzetta L, Rinaldi B, de Novellis V, Page CP, Barnes PJ, Cazzola M. Animal models of chronic obstructive pulmonary disease and their role in drug discovery and development: a critical review. Expert Opin Drug Discov 2025:1-20. [PMID: 39939153 DOI: 10.1080/17460441.2025.2466704] [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: 11/24/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION The use of laboratory animals is essential to understand the mechanisms underlying COPD and to discover and evaluate new drugs. However, the complex changes associated with the disease in humans are difficult to fully replicate in animal models. AREAS COVERED This review examines the most recent literature on animal models of COPD and their implications for drug discovery and development. EXPERT OPINION Recent advances in animal models include the introduction of transgenic mice with an increased propensity to develop COPD-associated features, such as emphysema, and animals exposed to relevant environmental agents other than cigarette smoke, in particular biomass smoke and other air pollutants. Other animal species, including zebrafish, pigs, ferrets and non-human primates, are also increasingly being used to gain insights into human COPD. Furthermore, three-dimensional organoids and humanized mouse models are emerging as technologies for evaluating novel therapeutics in more human-like models. However, despite these advances, no model has yet fully captured the heterogeneity and progression of COPD as observed in humans. Therefore, further research is needed to develop improved models incorporating humanized elements in experimental animals, that may better predict therapeutic responses in clinic settings and accelerate the development of new treatments for this debilitating disease.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Barbara Rinaldi
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Vito de Novellis
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Clive P Page
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Peter J Barnes
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
9
|
Cai Y, Wang Y, He Y, Ren K, Liu Z, Zhao L, Wei T. Utilizing alternative in vivo animal models for food safety and toxicity: A focus on thermal process contaminant acrylamide. Food Chem 2025; 465:142135. [PMID: 39579401 DOI: 10.1016/j.foodchem.2024.142135] [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: 07/06/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 11/25/2024]
Abstract
Rodent models have traditionally been used to assess the toxicity of food chemicals, but this approach is costly, time-consuming, and raises ethical concerns. Alternatively, non-mammalian models such as Drosophila melanogaster, Danio rerio, and Caenorhabditis elegans have been shown to be suitable for studying the toxicity of food hazards. Their advantages include low cost, short life cycles, adaptability to high-throughput screening, and adherence to the 3R principles of replacement, reduction, and refinement. These models have been extensively studied in the context of acrylamide toxicity, a common food contaminant. This article comprehensively reviews the biological characteristics of non-mammalian models, recent advances and challenges in acrylamide toxicity research using these models, and explores the potential of natural plant compounds in ameliorating acrylamide toxicity. The review aims to guide research using non-mammalian models for food safety assessment.
Collapse
Affiliation(s)
- Yang Cai
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yuhan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yanfei He
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Kefeng Ren
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Zongzhong Liu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Lingli Zhao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| | - Tian Wei
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| |
Collapse
|
10
|
Ford LM, Petersen-Jones SM. Modifiers and their impact on inherited retinal diseases: a review. Ophthalmic Genet 2025; 46:1-14. [PMID: 39780424 DOI: 10.1080/13816810.2024.2445221] [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: 06/06/2024] [Revised: 10/24/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The phenotypic variability of inherited conditions can be due to several factors including environmental, epigenetic, and genetic. One of those genetic factors is the presence of modifying loci which alter the phenotypic expression of a primary disease or phenotype-causing variant. Modifiers are known to affect penetrance, dominance, expressivity, and pleiotropy of disease. METHODS We review the literature to highlight the impact of modifiers on inherited retinal diseases. RESULTS Modifiers have been identified or associated with phenotypic variation in many inherited retinal diseases including retinitis pigmentosa and Stargardt disease. Despite being notoriously difficult to identify, proposed candidate modifiers have been identified using multiple methods including GWAS, family and population studies, and variant calling methods. CONCLUSIONS Overall, modifiers present themselves as an interesting target for further understanding of underlying disease pathways that could ultimately lead to therapeutic targets.
Collapse
Affiliation(s)
- Laura M Ford
- Genetics and Genome Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
11
|
Teitz-Tennenbaum S, Marinetti KN, Lahiri S, Siddiqui K, Flory C, Tennenbaum K, Hicks HG, Song B, Ganguly A, Osterholzer JJ. Sulfur dioxide exposure of mice induces peribronchiolar fibrosis-A defining feature of deployment-related constrictive bronchiolitis. PLoS One 2025; 20:e0313992. [PMID: 39854594 PMCID: PMC11761160 DOI: 10.1371/journal.pone.0313992] [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] [Received: 03/23/2023] [Accepted: 10/01/2024] [Indexed: 01/26/2025] Open
Abstract
Deployment-related constrictive bronchiolitis (DRCB) has emerged as a health concern in military personnel returning from Southwest Asia. Exposure to smoke from a fire at the Al-Mishraq sulfur enrichment facility and/or burn pits was reported by a subset of Veterans diagnosed with this disorder. DRCB is characterized by thickening and fibrosis of small airways (SA) in the lung, but whether these are related to toxin inhalation remains uncertain. The aim of this study was to determine whether sulfur dioxide (SO2) exposure can induce histopathological features of DRCB. C57BL/6J mice were exposed to 50 ± 5 ppm SO2 for one hour/day for five consecutive days. Lungs from exposed and unexposed mice were evaluated on day 5, 10, and 20. Lung sections were stained using hematoxylin and eosin, Masson's trichrome, picrosirius red (PSR), and immunofluorescence for club cell secretory protein, acetylated-α-tubulin, and Ki67. Small airway wall thickness was determined by morphometric analysis and collagen content was quantified by measuring PSR fluorescence intensity. CurveAlign and CT-FIRE were used to enumerate collagen fibers and assess fibers' width and length, respectively. Leukocyte subpopulations were quantified by flow cytometry analysis. This protocol of SO2 exposure of mice: 1) Triggered club cell proliferation and differentiation; 2) Increased SA wall thickness by inducing subepithelial collagen deposition; and 3) Increased width, length, and number, but not density, of collagen fibers within the wall of SA. 4) Induced no peribronchiolar inflammation or respiratory bronchiolitis. Collectively, these findings implicate club cell proliferation and differentiation in the profibrotic response to SO2 and identify this SO2 exposure as a potentially effective though imperfect model for studying SA fibrosis in DRCB.
Collapse
Affiliation(s)
- Seagal Teitz-Tennenbaum
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kayla N. Marinetti
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shayanki Lahiri
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Khadijah Siddiqui
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Celia Flory
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karinne Tennenbaum
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Helen G. Hicks
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Brian Song
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
| | - Anutosh Ganguly
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John J. Osterholzer
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
12
|
Obafemi OT, Ayeleso AO, Adewale OB, Unuofin J, Ekundayo BE, Ntwasa M, Lebelo SL. Animal models in biomedical research: Relevance of Drosophila melanogaster. Heliyon 2025; 11:e41605. [PMID: 39850441 PMCID: PMC11754520 DOI: 10.1016/j.heliyon.2024.e41605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Animal models have become veritable tools in gaining insight into the pathogenesis and progression of several human diseases. These models could range in complexity from Caenorhabditis elegans to non-human primates. With the aid of these animal models, a lot of new knowledge has been gained about several diseases which otherwise would not have been possible. Most times, the utilization of these animal models is predicated on the level of homology they share with humans, which suggests that outcomes of studies using them could be extrapolated to humans. However, this has not always been the case. Drosophila melanogaster is becoming increasingly relevant as preferred model for understanding the biochemical basis of several human diseases. Apart from its relatively short lifespan, high fecundity and ease of rearing, the simplicity of its genome and lower redundancy of its genes when compared with vertebrate models, as well as availability of genetic tool kit for easy manipulation of its genome, have all contributed to its emergence as a valid animal model of human diseases. This review aimed at highlighting the contributions of selected animal models in biomedical research with a focus on the relevance of Drosophila melanogaster in understanding the biochemical basis of some diseases that have continued to plague mankind.
Collapse
Affiliation(s)
- Olabisi Tajudeen Obafemi
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Ademola Olabode Ayeleso
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
- Biochemistry Programme, College of Agriculture, Engineering and Science, Bowen University, PMB 284, Iwo, Osun State, Nigeria
| | | | - Jeremiah Unuofin
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | | | - Monde Ntwasa
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Sogolo Lucky Lebelo
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| |
Collapse
|
13
|
Fucarino A, Pitruzzella A, Burgio S, Intili G, Manna OM, Modica MD, Poma S, Benfante A, Tomasello A, Scichilone N, Bucchieri F. A novel approach to investigate severe asthma and COPD: the 3d ex vivo respiratory mucosa model. J Asthma 2025; 62:110-123. [PMID: 39096201 DOI: 10.1080/02770903.2024.2388781] [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: 03/07/2024] [Revised: 06/23/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Purpose: This article illustrates the replication of asthma and COPD conditions in a laboratory setting and the potential applications of this methodology. Introduction: Biologic drugs have been shown to enhance the treatment of severe asthma and COPD. Monoclonal antibodies against specific targets have dramatically changed the management of these conditions. Although the inflammatory pathways of asthma and COPD have already been clearly outlined, alternative mechanisms of action remain mostly unexplored. They could provide additional insights into these diseases and their clinical management. Aims: In vivo or in vitro models have thus been developed to test alternative hypotheses. This study describes sophisticated ex vivo models that mimic the response of human respiratory mucosa to disease triggers, aiming to narrow the gap between laboratory studies and clinical practice. Results: These models successfully replicate crucial aspects of these diseases, such as inflammatory cell presence, cytokine production, and changes in tissue structure, offering a dynamic platform for investigating disease processes and evaluating potential treatments, such as monoclonal antibodies. The proposed models have the potential to enhance personalized medicine approaches and patient-specific treatments, helping to advance the understanding and management of respiratory diseases.
Collapse
Affiliation(s)
- Alberto Fucarino
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Stefano Burgio
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Giorgia Intili
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Olga Maria Manna
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Michele Domenico Modica
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
- Department of Otorhinolaryngology, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Salvatore Poma
- Department of Otorhinolaryngology, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Alida Benfante
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Alessandra Tomasello
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Nicola Scichilone
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| |
Collapse
|
14
|
Franco C, Rezzani R. Methods and Models for Studying Mycobacterium tuberculosis in Respiratory Infections. Int J Mol Sci 2024; 26:18. [PMID: 39795880 PMCID: PMC11719571 DOI: 10.3390/ijms26010018] [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: 11/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Respiratory infections, including tuberculosis, constitute a major global health challenge. Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading causes of mortality worldwide. The disease's complexity is attributed to Mtb's capacity to persist in latent states, evade host immune defenses, and develop resistance to antimicrobial treatments, posing significant challenges for diagnosis and therapy. Traditional models, such as animal studies and two-dimensional (2D) in vitro systems, often fail to accurately recapitulate human-specific immune processes, particularly the formation of granulomas-a defining feature of tubercular infection. These limitations underscore the need for more physiologically relevant models to study TB pathogenesis. Emerging three-dimensional (3D) in vitro systems, including organoids and lung-on-chip platforms, offer innovative approaches to mimic the structural and functional complexity of the human lung. These models enable the recreation of key aspects of the tubercular granulomas, such as cellular interactions, oxygen gradients, and nutrient limitations, thereby providing deeper insights into Mtb pathogenesis. This review aims to elucidate the advantages of 3D in vitro systems in bridging the translational gap between traditional experimental approaches and clinical applications. Particular emphasis is placed on their potential to address challenges related to genetic variability in both the host and pathogen, thereby advancing tubercular research and therapeutic development.
Collapse
Affiliation(s)
- Caterina Franco
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale–SISDO), 25123 Brescia, Italy
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
15
|
Baric TJ, Reneer ZB. Animal Models, Therapeutics, and Vaccine Approaches to Emerging and Re-Emerging Flaviviruses. Viruses 2024; 17:1. [PMID: 39861790 PMCID: PMC11769264 DOI: 10.3390/v17010001] [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: 11/13/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Flaviviruses are arthropod-borne viruses primarily transmitted through the mosquito Aedes aegypti or Culex genus of mosquitos. These viruses are predominantly found in tropical and subtropical regions of the world with their geographical spread predicted to increase as global temperatures continue to rise. These viruses cause a variety of diseases in humans with the most prevalent being caused by dengue, resulting in hemorrhagic fever and associated sequala. Current approaches for therapeutic control of flavivirus infections are limited, and despite recent advances, there are no approved drugs. Vaccines, available for a few circulating flaviviruses, still have limited potential for controlling contemporary and future outbreaks. Mouse models provide us with a valuable tool to test the effectiveness of drugs and vaccines, yet for many flaviviruses, well-established mouse models are lacking. In this review, we highlight the current state of flavivirus vaccines and therapeutics, as well as our current understanding of mouse models for various flaviviruses.
Collapse
Affiliation(s)
| | - Z. Beau Reneer
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3500, USA;
| |
Collapse
|
16
|
Mo K, Wang Y, Lu C, Li Z. Insight into the role of macrophages in periodontitis restoration and development. Virulence 2024; 15:2427234. [PMID: 39535076 PMCID: PMC11572313 DOI: 10.1080/21505594.2024.2427234] [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: 01/15/2024] [Revised: 08/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Periodontitis is one of the chronic diseases that have the greatest impact on human health, and it is associated with several other chronic diseases. Tissue damage associated with periodontitis is often connected with immune response. Immune cells are a crucial component of the human immune system and are directly involved in periodontitis during the inflammatory phase of the disease. Macrophages, as a key component of the immune system, are responsible for defence, antigen presentation and phagocytosis in healthy tissue. They are also closely linked to the development and resolution of periodontitis, through mechanisms such as macrophage polarization, pattern recognition receptors recognition, efferocytosis, and Specialized Pro-resolving Mediators (SPMs) production. Additionally, apoptosis and autophagy are also known to play a role in the recovery of periodontitis. This review aims to investigate the aforementioned mechanisms in more detail and identify novel therapeutic approaches for periodontitis.
Collapse
Affiliation(s)
- Keyin Mo
- School of Stomatology, Jinan University, Guangzhou, China
| | - Yijue Wang
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Chunting Lu
- Science and Education Office, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zejian Li
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Balough JL, Dipali SS, Velez K, Kumar TR, Duncan FE. Hallmarks of female reproductive aging in physiologic aging mice. NATURE AGING 2024; 4:1711-1730. [PMID: 39672896 DOI: 10.1038/s43587-024-00769-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
The female reproductive axis is one of the first organ systems to age, which has consequences for fertility and overall health. Here, we provide a comprehensive overview of the biological process of female reproductive aging across reproductive organs, tissues and cells based on research with widely used physiologic aging mouse models, and describe the mechanisms that underpin these phenotypes. Overall, aging is associated with dysregulation of the hypothalamic-pituitary-ovarian axis, perturbations of the ovarian stroma, reduced egg quantity and quality, and altered uterine morphology and function that contributes to reduced capacity for fertilization and impaired embryo development. Ultimately, these age-related phenotypes contribute to altered pregnancy outcomes and adverse consequences in offspring. Conserved mechanisms of aging, as well as those unique to the reproductive system, underlie these phenotypes. The knowledge of such mechanisms will lead to development of therapeutics to extend female reproductive longevity and support endocrine function and overall health.
Collapse
Affiliation(s)
- Julia L Balough
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| | - Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karen Velez
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca E Duncan
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA.
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
18
|
Tharmapalan V, Wagner W. Biomarkers for aging of blood - how transferable are they between mice and humans? Exp Hematol 2024; 140:104600. [PMID: 39128692 DOI: 10.1016/j.exphem.2024.104600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Aging significantly impacts the hematopoietic system, reducing its regenerative capacity and ability to restore homeostasis after stress. Mouse models have been invaluable in studying this process due to their shorter lifespan and the ability to explore genetic, treatment, and environmental influences on aging. However, not all aspects of aging are mirrored between species. This review compares three key aging biomarkers in the hematopoietic systems of mice and humans: myeloid bias, telomere attrition, and epigenetic clocks. Myeloid bias, marked by an increased fraction of myeloid cells and decreased lymphoid cells, is a significant aging marker in mice but is scarcely observed in humans after childhood. Conversely, telomere length is a robust aging biomarker in humans, whereas mice exhibit significantly different telomere dynamics, making telomere length less reliable in the murine system. Epigenetic clocks, based on DNA methylation changes at specific genomic regions, provide precise estimates of chronologic age in both mice and humans. Notably, age-associated regions in mice and humans occur at homologous genomic locations. Epigenetic clocks, depending on the epigenetic signatures used, also capture aspects of biological aging, offering powerful tools to assess genetic and environmental impacts on aging. Taken together, not all blood aging biomarkers are transferable between mice and humans. When using murine models to extrapolate human aging, it may be advantageous to focus on aging phenomena observed in both species. In conclusion, although mouse models offer significant insights, selecting appropriate biomarkers is crucial for translating findings to human aging.
Collapse
Affiliation(s)
- Vithurithra Tharmapalan
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical Faculty, Aachen, Germany
| | - Wolfgang Wagner
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical Faculty, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
19
|
Cherubini A, Della Torre S, Pelusi S, Valenti L. Sexual dimorphism of metabolic dysfunction-associated steatotic liver disease. Trends Mol Med 2024; 30:1126-1136. [PMID: 38890029 DOI: 10.1016/j.molmed.2024.05.013] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver condition. MASLD is a sexually dimorphic condition, with its development and progression influenced by sex chromosomes and hormones. Estrogens typically protect against, whereas androgens promote, MASLD. Therapeutic approaches for a sex-specific personalized medicine include estrogen replacement, androgen blockers, and novel drugs targeting hormonal pathways. However, the interactions between hormonal factors and inherited genetic variation impacts MASLD risk, necessitating more tailored therapies. Understanding sex disparities and the role of estrogens could improve MASLD interventions and management, whereas clinical trials addressing sex differences are crucial for advancing personalized treatment. This review explores the underappreciated impact of sexual dimorphism in MASLD and discusses the potential therapeutic application of sex-related hormones.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Serena Pelusi
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
20
|
Bernar A, Bauer M, Schirmer M, Streif W, Gebetsberger J. Murine vs. Human Osteoblast Responses to Coagulation and Inflammatory Factors: Reconsidering the Use of Animal Models in Hemophilia a Research. Biomedicines 2024; 12:2666. [PMID: 39767573 PMCID: PMC11726731 DOI: 10.3390/biomedicines12122666] [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: 09/30/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Hemophilia A is associated with frequent bleeding episodes, joint damage, and reduced bone mineral density (BMD). The role of coagulation factors and inflammatory cytokines on bone metabolism, particularly on osteoblast function, is of increasing interest. However, significant inter-species differences in bone remodeling raise concerns about the translatability of findings from murine models to human systems. This study aims to investigate the effects of human coagulation factors and cytokines on bone formation, focusing on inter-species differences in the cell viability and mineralization of murine and human osteoblasts. Methods: Murine MC3T3-E1 and human SaOs-2 osteoblasts were cultured in osteoblast differentiation medium supplemented with various coagulation factors (FVIII, vWF, vWF-FVIII, FIX, FX, thrombin, and FVIII-thrombin) or cytokines (IL-6, TNF-α). Cell viability was assessed at both two-week and three-week time points using the CCK-8 assay, and mineralization was evaluated via Alizarin red S staining. Results: Coagulation factors significantly enhanced cell viability in human osteoblasts but had no effects on the murine counterpart. FX inhibited mineralization in human cells, while murine cells showed no significant changes. TNF-α stimulated mineralization in murine osteoblasts but inhibited it in human cells, highlighting species-specific responses to inflammatory cytokines. Similar trends in response patterns were observed at two and three weeks, with greater consistency at the later time point. Conclusions: These findings reveal critical inter-species differences in osteoblast responses to coagulation factors and cytokines, raising questions about the validity of using murine models to study human bone metabolism. Future research must account for these differences to ensure that preclinical models accurately reflect human pathophysiology, particularly in the context of hemophilia A.
Collapse
Affiliation(s)
- Aline Bernar
- Department of Pediatrics I, Medical University Innsbruck, 6020 Innsbruck, Austria; (A.B.); (W.S.)
| | - Monika Bauer
- Department of Internal Medicine II, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.S.)
| | - Michael Schirmer
- Department of Internal Medicine II, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.S.)
| | - Werner Streif
- Department of Pediatrics I, Medical University Innsbruck, 6020 Innsbruck, Austria; (A.B.); (W.S.)
| | - Jennifer Gebetsberger
- Department of Pediatrics I, Medical University Innsbruck, 6020 Innsbruck, Austria; (A.B.); (W.S.)
| |
Collapse
|
21
|
Zhang X, Liang J, Zhang D, Wang L, Ye S. Unraveling Whole-Genome Sequence and Functional Characterization of P. megaterium PH3. Foods 2024; 13:3555. [PMID: 39593971 PMCID: PMC11593290 DOI: 10.3390/foods13223555] [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: 10/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Priestia megaterium (P. megaterium PH3) is an endophytic bacterium isolated from peanuts. It has natural resveratrol production ability and shows potential application value. This study analyzed its genetic function and metabolic mechanism through whole-genome sequencing and found that the genome size is 5,960,365 bp, the GC content is 37.62%, and 6132 genes are annotated. Functional analysis showed that this strain contained 149 carbohydrate active enzyme genes, 7 secondary metabolite synthesis gene clusters, 509 virulence genes, and 273 drug-resistance genes. At the same time, this strain has the ability to regulate salt stress, low temperature, and hypoxia. Genomic analysis reveals a stilbene-synthase-containing type III polyketide synthase gene cluster that contributes to resveratrol synthesis. A safety assessment showed that the strain is non-hemolytic, does not produce amino acid decarboxylase, and is not resistant to multiple antibiotics. In the mouse model, P. megaterium PH3 did not have significant effects on body weight, behavior, or physiological indicators. These results provide important basic data and theoretical support for its industrial application and the research and development of plant protection agents.
Collapse
Affiliation(s)
- Xiaohan Zhang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.Z.); (J.L.); (D.Z.); (L.W.)
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, China
| | - Junbo Liang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.Z.); (J.L.); (D.Z.); (L.W.)
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, China
| | - Dong Zhang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.Z.); (J.L.); (D.Z.); (L.W.)
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, China
| | - Liang Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.Z.); (J.L.); (D.Z.); (L.W.)
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, China
| | - Shuhong Ye
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian 116034, China
| |
Collapse
|
22
|
Kaur G, Wang Q, Tjitropranoto A, Unwalla H, Rahman I. Cold ischemia time alters cell-type specific senescence leading to loss of cellular integrity in mouse lungs. Exp Lung Res 2024; 50:184-198. [PMID: 39427288 PMCID: PMC11513191 DOI: 10.1080/01902148.2024.2414974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/11/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Purpose: Ischemia-reperfusion injury (IRI) is a major challenge in lung transplantation often causing graft dysfunction and chronic airway illnesses in recipients. To prevent potential transplant related complications, strict guidelines were put in place to choose viable donor lungs with minimal risk of IRI. These regulations deem most of the donor organs unfit for transplant which then are donated for research to understand the mechanisms of health and diseases in human. However, resected organs that are being transported undergo cold ischemia that can negatively affect the tissue architecture and other cellular functions under study. Thus, it is important to assess how cold ischemia time (CIT) affects the physiological mechanism. In this respect, we are interested in studying how CIT affects cellular senescence in normal aging and various pulmonary pathologies. We thus hypothesized that prolonged CIT exhibits cell-type specific changes in lung cellular senescence in mice. Methods: Lung lobes from C57BL/6J (n = 5-8) mice were harvested and stored in UW Belzer cold storage solution for 0, 4-, 9-, 12-, 24-, and 48-h CIT. Lung cellular senescence was determined using fluorescence (C12FdG) assay and co-immunolabelling was performed to identify changes in individual cell types. Results: We found a rapid decline in the overall lung cellular senescence after 4-h of CIT in our study. Co-immunolabelling revealed the endothelial cells to be most affected by cold ischemia, demonstrating significant decrease in the endothelial cell senescence immediately after harvest. Annexin V-PI staining further revealed a prominent increase in the number of necrotic cells at 4-h CIT, thus suggesting that most of the cells undergo cell death within a few hours of cold ischemic injury. Conclusions: We thus concluded that CIT significantly lowers the cellular senescence in lung tissues and must be considered as a confounding factor for mechanistic studies in the future.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Ariel Tjitropranoto
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Hoshang Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
23
|
Lejri I, Cader Z, Grimm A, Eckert A. Human iPSCs from Aged Donors Retain Their Mitochondrial Aging Signature. Int J Mol Sci 2024; 25:11199. [PMID: 39456998 PMCID: PMC11508692 DOI: 10.3390/ijms252011199] [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: 09/25/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Aging represents the leading risk factor for developing neurodegenerative disorders. One of the nine hallmarks of aging is mitochondrial dysfunction. Age-related mitochondrial alterations have been shown to affect mitochondrial energy metabolism, reduction-oxidation homeostasis, and mitochondrial dynamics. Previous reports have shown that induced pluripotent stem cells (iPSCs) from aged donors do not keep the aging signature at the transcriptomic level. However, not all aspects of aging have been investigated, and especially not the mitochondria-related aging signature. Therefore, the present study compared the mitochondrial function in iPSCs from healthy aged donors compared to those of young donors. We addressed whether aged iPSCs may be used as drug-screening models of "aging in a dish" to identify therapies alleviating mitochondria aging. Compared to iPSCs from young donors, we demonstrate that iPSCs from aged donors show impaired mitochondrial bioenergetics and exhibit a rise in reactive oxygen species generation. Furthermore, aged iPSCs present a lower mitochondrial mass and alterations in the morphology of the mitochondrial network when compared to iPSCs from young donors. This study provides the first evidence that the aging phenotype is present at the mitochondrial level in iPSCs from aged donors, ranging from bioenergetics to mitochondrial network morphology. This model might be used to screen mitochondria-targeting drugs to promote healthy aging at the mitochondrial level.
Collapse
Affiliation(s)
- Imane Lejri
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| | - Zameel Cader
- Translational Molecular Neuroscience Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK;
| | - Amandine Grimm
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Department of Biomedicine, University of Basel, 4055 Basel, Switzerland
| | - Anne Eckert
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| |
Collapse
|
24
|
Selitrennikoff CP, Sylvia C, Sanchez M, Lawrence P, Trosch K, Carenza A, Meschter C. Evaluate the safety of a novel photohydrolysis technology used to clean and disinfect indoor air: A murine study. PLoS One 2024; 19:e0307031. [PMID: 39383125 PMCID: PMC11463749 DOI: 10.1371/journal.pone.0307031] [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] [Received: 03/04/2024] [Accepted: 06/27/2024] [Indexed: 10/11/2024] Open
Abstract
There is a pressing need to develop new technologies that continuously eliminates harmful pollutants and pathogens in occupied indoor spaces without compromising safety. This study was undertaken to test the safety of a novel air cleaning and disinfection technology called Advanced Photohydrolysis. Advanced Photohydrolysis generates a complex mixture of ions and molecules that are released into the air and has been shown to reduce airborne and surface pathogens. Mice (6-8-week-old) were exposed to therapeutic levels of Advanced Photohydrolysis for 90-days. During the study, the Advanced-Photohydrolysis-exposed and control mice were monitored for food consumption, body weight gain, and any overt adverse effects. In addition, at the conclusion of the study, the blood chemistry and hematology values of both groups were determined. Finally, the tissues of the conduction and respiratory portions of the airways of mice from both groups were examined for any pathological changes. The mice of both groups were found to be normal and healthy throughout the 90-day study; there were no differences in the behavior, food consumption and weight gain. Analysis of clinical chemistry values found no differences in hepatocellular function or other markers of cellular and organ function, and clinical hematology values were also unremarkable. Finally, and importantly, histopathology of the upper and lower airway tissues showed no deleterious effects. These results are the first to demonstrate directly the safety of Advanced Photohydrolysis on live mammals and encourage additional studies.
Collapse
Affiliation(s)
- Claude P. Selitrennikoff
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles Sylvia
- Comparative Biosciences, Inc., Sunnyvale, California, United States of America
| | - Maria Sanchez
- Comparative Biosciences, Inc., Sunnyvale, California, United States of America
| | | | - Kimberly Trosch
- ActivePure Technologies, Dallas, Texas, United States of America
| | - Amy Carenza
- ActivePure Technologies, Dallas, Texas, United States of America
| | - Carol Meschter
- Comparative Biosciences, Inc., Sunnyvale, California, United States of America
| |
Collapse
|
25
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
26
|
Iosageanu A, Stefan LM, Craciunescu O, Cimpean A. Anti-Inflammatory and Wound Healing Properties of Different Honey Varieties from Romania and Correlations to Their Composition. Life (Basel) 2024; 14:1187. [PMID: 39337969 PMCID: PMC11432766 DOI: 10.3390/life14091187] [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: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The complex composition of honey plays a crucial role in wound healing, exhibiting varying effects at different stages of the healing process. This study investigated seven honey varieties sourced from different regions of Romania using in vitro experimental models developed in macrophage-like, fibroblast, and keratinocyte cell lines to explore the mechanisms by which honey promoted the healing process. This study assessed the impact of honey on inflammatory cytokine production in macrophage-like cells, cell proliferation and collagen synthesis in fibroblasts, and cell proliferation and migration in keratinocytes. Additionally, correlation analysis was conducted to examine the relationship between honey composition and its biological properties. Honey varieties presented both anti- and pro-inflammatory effects. Moreover, they displayed dose-dependent pro-proliferative effects, stimulating collagen synthesis and cell migration, thereby enhancing the re-epithelialization process. The Pearson coefficient analysis indicated a strong positive correlation between biological activities and phenolic content. Additionally, there was a medium positive correlation with the ascorbic acid content and a medium negative correlation with the glucose content in the different honey varieties. Romanian honey varieties rich in phenolics showed potential in modulating inflammation, proliferation, collagen synthesis, and cell migration, suggesting their suitability for further evaluation and development of innovative dressings for skin tissue regeneration.
Collapse
Affiliation(s)
- Andreea Iosageanu
- Faculty of Biology, University of Bucharest, 91-95, Splaiul Independentei, 050095 Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Laura Mihaela Stefan
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Oana Craciunescu
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Anisoara Cimpean
- Faculty of Biology, University of Bucharest, 91-95, Splaiul Independentei, 050095 Bucharest, Romania
| |
Collapse
|
27
|
Liu SX, Harris AC, Gewirtz JC. How life events may confer vulnerability to addiction: the role of epigenetics. Front Mol Neurosci 2024; 17:1462769. [PMID: 39359689 PMCID: PMC11446245 DOI: 10.3389/fnmol.2024.1462769] [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: 07/10/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024] Open
Abstract
Substance use disorder (SUD) represents a large and growing global health problem. Despite the strong addictive potency of drugs of abuse, only a minority of those exposed develop SUDs. While certain life experiences (e.g., childhood trauma) may increase subsequent vulnerability to SUDs, mechanisms underlying these effects are not yet well understood. Given the chronic and relapsing nature of SUDs, and the length of time that can elapse between prior life events and subsequent drug exposure, changes in SUD vulnerability almost certainly involve long-term epigenetic dysregulation. To validate this idea, functional effects of specific epigenetic modifications in brain regions mediating reinforcement learning (e.g., nucleus accumbens, prefrontal cortex) have been investigated in a variety of animal models of SUDs. In addition, the effects of epigenetic modifications produced by prior life experiences on subsequent SUD vulnerability have been studied, but mostly in a correlational manner. Here, we review how epigenetic mechanisms impact SUD-related behavior in animal models and summarize our understanding of the relationships among life experiences, epigenetic regulation, and future vulnerability to SUDs. Despite variations in study design, epigenetic modifications that most consistently affect SUD-related behavior are those that produce predominantly unidirectional effects on gene regulation, such as DNA methylation and histone phosphorylation. Evidence explicitly linking environmentally induced epigenetic modifications to subsequent SUD-related behavior is surprisingly sparse. We conclude by offering several directions for future research to begin to address this critical research gap.
Collapse
Affiliation(s)
- Shirelle X Liu
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | - Andrew C Harris
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Hennepin Healthcare Research Institute, Minneapolis, MN, United States
| | - Jonathan C Gewirtz
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
28
|
Dang J, Cai T, Tuo Y, Peng S, Wang J, Gu A, Li J, Ding L, Du S, Wang L. Corn Peptides Alleviate Nonalcoholic Fatty Liver Fibrosis in Mice by Inhibiting NLRP3 Inflammasome Activation and Regulating Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19378-19394. [PMID: 39166383 DOI: 10.1021/acs.jafc.4c04232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
This study aimed to investigate the effects of corn gluten-derived soluble epoxide hydrolase (sEH) inhibitory peptides on nonalcoholic fatty liver fibrosis induced by a high-fat diet and carbon tetrachloride in mice. Mice treated with corn peptides at doses of 500 or 1000 mg/kg/d for 4 weeks exhibited reduced sEH activity in serum and liver, enhanced lipid metabolism, and decreased lipid accumulation and oxidative stress. Corn peptides effectively downregulated the mRNA levels of Pro-IL-1β, Pro-IL-18, NOD-like receptor protein 3 (NLRP3), ASC, Pro-caspase-1, Caspase-1, and GSDMD in the liver. This hepatoprotective effect of corn peptides by inhibiting NLRP3 inflammasome activation was further validated in H2O2-induced HepG2 cells. Moreover, corn peptides restored the composition of the gut microbiota and promoted short-chain fatty acid production. This study provides evidence that corn-derived sEH inhibitory peptides have hepatoprotective activity against nonalcoholic fatty liver fibrosis by suppressing NLRP3 inflammasome activation and modulating gut microbiota.
Collapse
Affiliation(s)
- Jiamin Dang
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Ting Cai
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Yuanrong Tuo
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Siwang Peng
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Jing Wang
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Aiying Gu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Jialu Li
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Long Ding
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Shuangkui Du
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
- Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Xianyang, Shaanxi Province 712100, People's Republic of China
| | - Liying Wang
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi Province 712100, People's Republic of China
- Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Xianyang, Shaanxi Province 712100, People's Republic of China
| |
Collapse
|
29
|
Gopallawa I, Gupta C, Jawa R, Cyril A, Jawa V, Chirmule N, Gujar V. Applications of Organoids in Advancing Drug Discovery and Development. J Pharm Sci 2024; 113:2659-2667. [PMID: 39002723 DOI: 10.1016/j.xphs.2024.06.016] [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: 03/28/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
Organoids are small, self-organizing three-dimensional cell cultures that are derived from stem cells or primary organs. These cultures replicate the complexity of an organ, which cannot be achieved by single-cell culture systems. Organoids can be used in testing of new drugs instead of animals. Development and validation of organoids is thus important to reduce the reliance on animals for drug testing. In this review, we have discussed the developmental and regulatory aspects of organoids and highlighted their importance in drug development. We have first summarized different types of culture-based organoid systems such as submerged Matrigel, micro-fluidic 3D cultures, inducible pluripotent stem cells, and air-liquid interface cultures. These systems help us understand the intricate interplay between cells and their surrounding milieu for identifying functions of target receptors, soluble factors, and spatial interactions. Further, we have discussed the advances in humanized severe-combined immunodeficiency mouse models and their applications in the pharmacology of immune-oncology. Since regulatory aspects are important in using organoids for drug development, we have summarized FDA and EMA regulations on organoid research to support pre-clinical studies. Finally, we have included some unique studies highlighting the use of organoids in studying infectious diseases, cancer, and fundamental biology. These studies also exemplify the latest technological advances in organoid development resulting in improved efficiency. Overall, this review comprehensively summarizes the applications of organoids in early drug development during discovery and pre-clinical studies.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | | | - Rayan Jawa
- University of Pennsylvania, Philadelphia, PA, USA
| | - Arya Cyril
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, NY, USA.
| | | | - Vikramsingh Gujar
- Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| |
Collapse
|
30
|
Stańczyk M, Szubart N, Maslanka R, Zadrag-Tecza R. Mitochondrial Dysfunctions: Genetic and Cellular Implications Revealed by Various Model Organisms. Genes (Basel) 2024; 15:1153. [PMID: 39336744 PMCID: PMC11431519 DOI: 10.3390/genes15091153] [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: 07/30/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondria play a crucial role in maintaining the energy status and redox homeostasis of eukaryotic cells. They are responsible for the metabolic efficiency of cells, providing both ATP and intermediate metabolic products. They also regulate cell survival and death under stress conditions by controlling the cell response or activating the apoptosis process. This functional diversity of mitochondria indicates their great importance for cellular metabolism. Hence, dysfunctions of these structures are increasingly recognized as an element of the etiology of many human diseases and, therefore, an extremely promising therapeutic target. Mitochondrial dysfunctions can be caused by mutations in both nuclear and mitochondrial DNA, as well as by stress factors or replication errors. Progress in knowledge about the biology of mitochondria, as well as the consequences for the efficiency of the entire organism resulting from the dysfunction of these structures, is achieved through the use of model organisms. They are an invaluable tool for analyzing complex cellular processes, leading to a better understanding of diseases caused by mitochondrial dysfunction. In this work, we review the most commonly used model organisms, discussing both their advantages and limitations in modeling fundamental mitochondrial processes or mitochondrial diseases.
Collapse
Affiliation(s)
| | | | | | - Renata Zadrag-Tecza
- Institute of Biology, College of Natural Sciences, University of Rzeszow, 35-959 Rzeszow, Poland; (M.S.); (N.S.); (R.M.)
| |
Collapse
|
31
|
Hood MN, Ayompe E, Holmes-Hampton GP, Korotcov A, Wuddie K, Aschenake Z, Ahmed AE, Creavalle M, Knollmann-Ritschel B. Preliminary Promising Findings for Manganese Chloride as a Novel Radiation Countermeasure Against Acute Radiation Syndrome. Mil Med 2024; 189:598-607. [PMID: 39160887 DOI: 10.1093/milmed/usae198] [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: 11/26/2023] [Revised: 02/01/2024] [Accepted: 04/05/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Military members and first responders may, at moment's notice, be asked to assist in incidents that may result in radiation exposure such as Operation Tomadachi in which the U.S. Navy provided significant relief for the Fukushima Daiichi Nuclear Reactor accident in Japan after an earthquake and tsunami in 2011. We are also currently facing potential threats from nuclear power plants in the Ukraine should a power disruption to a nuclear plant interfere with cooling or other safety measures. Exposure to high doses of radiation results in acute radiation syndrome (ARS) characterized by symptoms arising from hematopoietic, gastrointestinal, and neurovascular injuries. Although there are mitigators FDA approved to treat ARS, there are currently no FDA-approved prophylactic medical interventions to help protect persons who may need to respond to radiation emergencies. There is strong evidence that manganese (Mn) has radiation protective efficacy as a promising prophylactic countermeasure. MATERIALS AND METHODS All animal procedures were approved by the Institutional Animal Care and Use Committee. Male and female B6D2F1J mice, 10 to 11 weeks old, were used for neurotoxicity studies and temporal effects of Mn. Four groups were evaluated: (1) vehicle injection, (2) dose of 4.5 mg/kg for 3 days, (3) dose of 13.5 mg/kg, and (4) sham. Irradiated mice were exposed to 9.5 Gy whole body Co60 γ-radiation. MRI was performed with a high dose of manganese chloride (MnCl2) (150 mg/kg) to assess the distribution of the MnCl2. RESULTS The mice have promising survival curves (highest survival-13.5 mg/kg dose over 3 days of MnCl2 at 80% [87% female, 73% male] P = 0.0004). The complete blood count (CBC) results demonstrated a typical hematopoietic response in all of the irradiated groups, followed by mildly accelerated recovery by day 28 in the treated groups. No difference between groups was measured by Rota Rod, DigiGait, and Y-maze. Histologic evaluation of the bone marrow sections in the group given 13.5 mg/kg dose over 3 days had the best return to cellularity at 80%. MRI showed a systemic distribution of MnCl2. DISCUSSION The preliminary data suggest that a dose of 13.5 mg/kg of MnCl2 given over 3 days prior to exposure of radiation may have a protective benefit while not exhibiting the neurobehavioral problems. A countermeasure that can prophylactically protect emergency personnel entering an area contaminated with high levels of radiation is needed, especially in light that nuclear accidents are a continued global threat. There is a need for a protective agent with easy long-term storage, easy to transport, easy to administer, and low cost. Histologic evaluation supports the promising effect of MnCl2 in protecting tissue, especially the bone marrow using the dose given over 3 days (4.5 mg/kg per day) of MnCl2. CONCLUSIONS Initial experiments show that MnCl2 is a promising safe and effective prophylactic countermeasure against ARS. MRI data support the systemic distribution of MnCl2 which is needed in order to protect multiple tissues in the body. The pathology data in bone marrow and the brain support faster recovery from radiation exposure in the treated animals and decreased organ damage.
Collapse
Affiliation(s)
- Maureen N Hood
- Department of Radiology & Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Emmanuel Ayompe
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Gregory P Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Alexandru Korotcov
- Department of Radiology & Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Kefale Wuddie
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Zemenu Aschenake
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Anwar E Ahmed
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Marqus Creavalle
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | |
Collapse
|
32
|
Duncan RP, Moustafa DA, Lewin GR, Diggle FL, Bomberger JM, Whiteley M, Goldberg JB. Improvement of a mouse infection model to capture Pseudomonas aeruginosa chronic physiology in cystic fibrosis. Proc Natl Acad Sci U S A 2024; 121:e2406234121. [PMID: 39102545 PMCID: PMC11331117 DOI: 10.1073/pnas.2406234121] [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: 03/26/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Laboratory models are central to microbiology research, advancing the understanding of bacterial physiology by mimicking natural environments, from soil to the human microbiome. When studying host-bacteria interactions, animal models enable investigators to examine bacterial dynamics associated with a host, and in the case of human infections, animal models are necessary to translate basic research into clinical treatments. Efforts toward improving animal infection models are typically based on reproducing host genotypes/phenotypes and disease manifestations, leaving a gap in how well the physiology of microbes reflects their behavior in a human host. Understanding bacterial physiology is vital because it dictates host response and bacterial interactions with antimicrobials. Thus, our goal was to develop an animal model that accurately recapitulates bacterial physiology in human infection. The system we chose to model was a chronic Pseudomonas aeruginosa respiratory infection in cystic fibrosis (CF). To accomplish this goal, we leveraged a framework that we recently developed to evaluate model accuracy by calculating the percentage of bacterial genes that are expressed similarly in a model to how they are expressed in their infection environment. We combined two complementary models of P. aeruginosa infection-an in vitro synthetic CF sputum model (SCFM2) and a mouse acute pneumonia model. This combined model captured the chronic physiology of P. aeruginosa in CF better than the standard mouse infection model, showing the power of a data-driven approach to refining animal models. In addition, the results of this work challenge the assumption that a chronic infection model requires long-term colonization.
Collapse
Affiliation(s)
- Rebecca P. Duncan
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| | - Dina A. Moustafa
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| | - Gina R. Lewin
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Frances L. Diggle
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Jennifer M. Bomberger
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA15219
| | - Marvin Whiteley
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Joanna B. Goldberg
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| |
Collapse
|
33
|
Singh N, Nagar E, Roy D, Arora N. NLRP3/GSDMD mediated pyroptosis induces lung inflammation susceptibility in diesel exhaust exposed mouse strains. Gene 2024; 918:148459. [PMID: 38608794 DOI: 10.1016/j.gene.2024.148459] [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: 12/14/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Genetic diversity among species influences the disease severity outcomes linked to air pollution. However, the mechanism responsible for this variability remain elusive and needs further investigation. OBJECTIVE To investigate the genetic factors and pathways linked with differential susceptibility in mouse strains associated with diesel exhaust exposure. METHODS C57BL/6 and Balb/c mice were exposed to diesel exhaust (DE) for 5 days/week for 30 min/day for 8 weeks. Body weight of mice was recorded every week and airway hyperresponsiveness towards DE exposure was recorded after 24 h of last exposure. Mice were euthanised to collect BALF, blood, lung tissues for immunobiochemical assays, structural integrity and genetic studies. RESULTS C57BL/6 mice showed significantly decreased body weight in comparison to Balb/c mice (p < 0.05). Both mouse strains showed lung resistance and damage to elastance upon DE exposure compared to respective controls (p < 0.05) with more pronounced effects in C57BL/6 mice. Lung histology showed increase in bronchiolar infiltration and damage to the wall in C57BL/6 mice (p < 0.05). DE exposure upregulated pro-inflammatory and Th2 cytokine levels in C57BL/6 in comparison to Balb/c mice. C57BL/6 mice showed increase in Caspase-1 and ASC expression confirming activation of downstream pathway. This showed significant activation of inflammasome pathway in C57BL/6 mice with ∼2-fold increase in NLRP3 and elevated IL-1β expression. Gasdermin-D levels were increased in C57BL/6 mice demonstrating induction of pyroptosis that corroborated with IL-1β secretion (p < 0.05). Genetic variability among both species was confirmed with sanger's sequencing suggesting presence of SNPs in 3'UTRs of IL-1β gene influencing expression between mouse strains. CONCLUSIONS C57BL/6 mice exhibited increased susceptibility to diesel exhaust in contrast to Balb/c mice via activation of NLRP3-related pyroptosis. Differential susceptibility between strains may be attributed via SNPs in the 3'UTRs of the IL-1β gene.
Collapse
Affiliation(s)
- Naresh Singh
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ekta Nagar
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepti Roy
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Naveen Arora
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
34
|
Kim SY, Choi JA, Choi S, Kim KK, Song CH, Kim EM. Advances in an In Vitro Tuberculosis Infection Model Using Human Lung Organoids for Host-Directed Therapies. PLoS Pathog 2024; 20:e1012295. [PMID: 39052544 PMCID: PMC11271890 DOI: 10.1371/journal.ppat.1012295] [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] [Received: 11/17/2023] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
The emergence of drug-resistant Mycobacterium tuberculosis (M.tb) has led to the development of novel anti-tuberculosis (anti-TB) drugs. Common methods for testing the efficacy of new drugs, including two-dimensional cell culture models or animal models, have several limitations. Therefore, an appropriate model representative of the human organism is required. Here, we developed an M.tb infection model using human lung organoids (hLOs) and demonstrated that M.tb H37Rv can infect lung epithelial cells and human macrophages (hMφs) in hLOs. This novel M.tb infection model can be cultured long-term and split several times while maintaining a similar number of M.tb H37Rv inside the hLOs. Anti-TB drugs reduced the intracellular survival of M.tb in hLOs. Notably, M.tb growth in hLOs was effectively suppressed at each passage by rifampicin and bedaquiline. Furthermore, a reduction in inflammatory cytokine production and intracellular survival of M.tb were observed upon knockdown of MFN2 and HERPUD1 (host-directed therapeutic targets for TB) in our M.tb H37Rv-infected hLO model. Thus, the incorporation of hMφs and M.tb into hLOs provides a powerful strategy for generating an M.tb infection model. This model can effectively reflect host-pathogen interactions and be utilized to test the efficacy of anti-TB drugs and host-directed therapies.
Collapse
Affiliation(s)
- Seung-Yeon Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University Daejeon, Republic of Korea
| | - Ji-Ae Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Seri Choi
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Kee K. Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University Daejeon, Republic of Korea
| | - Chang-Hwa Song
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Mi Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
- Department of Bio & Environmental Technology, College of Science and Convergence Technology, Seoul Women’s University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Guo J, Zhao Y, Kang SG, Huang K, Tong T. Differential effects of four laboratory animal control diets on gut microbiota in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4438-4452. [PMID: 38323712 DOI: 10.1002/jsfa.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND The gut microbiota is intricate and susceptible to multiple factors, with diet being a major contributor. The present study aimed to investigate the impact of four commonly used laboratory animal control diets, namely Keao Xieli's maintenance diet (KX), HFK's 1025 (HF), Research Diets' D12450B (RD), and Lab Diet's 5CC4 (LD), on the gut microbiota of mice. RESULTS A total of 40 mice were randomly assigned to four groups, and each group was fed one of the four diets for a duration of 8 weeks. The assessment of gut microbiota was conducted using 16S rRNA sequencing both at the beginning of the study (week 0) and the end (week 8), which served as the baseline and endpoint samples, respectively. Following the 8-week feeding period, no significant differences were observed in physiological parameters, including body weight, visceral weight, and blood biochemical indices, across the four groups. Nonetheless, relative to the baseline, discernible alterations in the gut microbiota were observed in all groups, encompassing shifts in beta-diversity, hierarchical clustering, and key genera. Among the four diets, HF diet exhibited a significant influence on alpha-diversity, RD diet brought about notable changes in microbial composition at the phylum level, and LD diet demonstrated an interconnected co-occurrence network. Mantel analysis indicated no significant correlation between physiological parameters and gut microbiota in the four groups. CONCLUSION Overall, our study demonstrated that the four control diets had a minimal impact on physiological parameters, while exerting a distinct influence on the gut microbiota after 8 weeks. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Yuhan Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, P. R. China
- Beijing Laboratory for Food Quality and Safety, Beijing, P. R. China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, P. R. China
- Beijing Laboratory for Food Quality and Safety, Beijing, P. R. China
| |
Collapse
|
36
|
Jia J, Zhang M, Cao Z, Hu X, Lei S, Zhang Y, Kang X. The rabbit model for spinal tuberculosis: An overview. J Orthop Surg (Hong Kong) 2024; 32:10225536241266703. [PMID: 39033332 DOI: 10.1177/10225536241266703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Mycobacterium tuberculosis infection has emerged as a global public health issue, predominantly manifesting as pulmonary tuberculosis. Bone and joint tuberculosis, with spinal tuberculosis accounting for approximately 50%, represents a significant form of extrapulmonary tuberculosis. Over the past years, there has been a rise in the incidence of spinal tuberculosis, and research concerning this area has gained significant attention. At present, animal models provide a means to investigate the pathogenesis, drug resistance, and novel treatment approaches for spinal tuberculosis. New Zealand rabbits, possessing a comparable anatomical structure to humans and capable of reproducing typical pathological features of human tuberculosis, are extensively employed in spinal tuberculosis research using animal models. This article comprehensively evaluates the strengths, considerations in strain selection, various modelling approaches, and practical applications of the rabbit model in studying spinal tuberculosis based on pertinent literature to guide fundamental research in this field by providing valuable insights into appropriate animal model selection.
Collapse
Affiliation(s)
- Jingwen Jia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Mingtao Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Zhenyu Cao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Xuchang Hu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Shuanhu Lei
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Yizhi Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Xuewen Kang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| |
Collapse
|
37
|
Cogut V, Goris M, Jansma A, van der Staaij M, Henning RH. Hippocampal neuroimmune response in mice undergoing serial daily torpor induced by calorie restriction. Front Neuroanat 2024; 18:1334206. [PMID: 38686173 PMCID: PMC11056553 DOI: 10.3389/fnana.2024.1334206] [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: 11/06/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
Hibernating animals demonstrate a remarkable ability to withstand extreme physiological brain changes without triggering adverse neuroinflammatory responses. While hibernators may offer valuable insights into the neuroprotective mechanisms inherent to hibernation, studies using such species are constrained by the limited availability of molecular tools. Laboratory mice may serve as an alternative, entering states of hypometabolism and hypothermia similar to the torpor observed in hibernation when faced with energy shortage. Notably, prolonged calorie restriction (CR) induces serial daily torpor patterns in mice, comparable to species that utilize daily hibernation. Here, we examined the neuroinflammatory response in the hippocampus of male C57BL/6 mice undergoing serial daily torpor induced by a 30% CR for 4 weeks. During daily torpor episodes, CR mice exhibited transient increases in TNF-α mRNA expression, which normalized upon arousal. Concurrently, the CA1 region of the hippocampus showed persistent morphological changes in microglia, characterized by reduced cell branching, decreased cell complexity and altered shape. Importantly, these morphological changes were not accompanied by evident signs of astrogliosis or oxidative stress, typically associated with detrimental neuroinflammation. Collectively, the adaptive nature of the brain's inflammatory response to CR-induced torpor in mice parallels observations in hibernators, highlighting its value for studying the mechanisms of brain resilience during torpor. Such insights could pave the way for novel therapeutic interventions in stroke and neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
38
|
Vo Q, Benam KH. Advancements in preclinical human-relevant modeling of pulmonary vasculature on-chip. Eur J Pharm Sci 2024; 195:106709. [PMID: 38246431 PMCID: PMC10939731 DOI: 10.1016/j.ejps.2024.106709] [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: 11/05/2023] [Revised: 01/05/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Preclinical human-relevant modeling of organ-specific vasculature offers a unique opportunity to recreate pathophysiological intercellular, tissue-tissue, and cell-matrix interactions for a broad range of applications. Lung vasculature is particularly important due to its involvement in genesis and progression of rare, debilitating disorders as well as common chronic pathologies. Here, we provide an overview of the latest advances in the development of pulmonary vascular (PV) models using emerging microfluidic tissue engineering technology Organs-on-Chips (so-called PV-Chips). We first review the currently reported PV-Chip systems and their key features, and then critically discuss their major limitations in reproducing in vivo-seen and disease-relevant cellularity, localization, and microstructure. We conclude by presenting latest efforts to overcome such technical and biological limitations and future directions.
Collapse
Affiliation(s)
- Quoc Vo
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kambez H Benam
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
39
|
Sumi M, Westcott R, Stuehr E, Ghosh C, Stuehr DJ, Ghosh A. Regional variations in allergen-induced airway inflammation correspond to changes in soluble guanylyl cyclase heme and expression of heme oxygenase-1. FASEB J 2024; 38:e23572. [PMID: 38512139 PMCID: PMC10977653 DOI: 10.1096/fj.202301626rrr] [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: 08/09/2023] [Revised: 02/09/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Asthma is characterized by airway remodeling and hyperreactivity. Our earlier studies determined that the nitric oxide (NO)-soluble guanylyl cyclase (sGC)-cGMP pathway plays a significant role in human lung bronchodilation. However, this bronchodilation is dysfunctional in asthma due to high NO levels, which cause sGC to become heme-free and desensitized to its natural activator, NO. In order to determine how asthma impacts the various lung segments/lobes, we mapped the inflammatory regions of lungs to determine whether such regions coincided with molecular signatures of sGC dysfunction. We demonstrate using murine models of asthma (OVA and CFA/HDM) that the inflamed segments of these murine lungs can be tracked by upregulated expression of HO1 and these regions in turn overlap with regions of heme-free sGC as evidenced by a decreased sGC-α1β1 heterodimer and an increased response to heme-independent sGC activator, BAY 60-2770, relative to naïve uninflamed regions. We also find that NO generated from iNOS upregulation in the inflamed segments has a higher impact on developing heme-free sGC as increasing iNOS activity correlates linearly with elevated heme-independent sGC activation. This excess NO works by affecting the epithelial lung hemoglobin (Hb) to become heme-free in asthma, thereby causing the Hb to lose its NO scavenging function and exposing the underlying smooth muscle sGC to excess NO, which in turn becomes heme-free. Recognition of these specific lung segments enhances our understanding of the inflamed lungs in asthma with the ultimate aim to evaluate potential therapies and suggest that regional and not global inflammation impacts lung function in asthma.
Collapse
Affiliation(s)
- Mamta Sumi
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| | - Rosemary Westcott
- Department of Biomedical Engineering, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| | - Eric Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| | - Chaitali Ghosh
- Department of Biomedical Engineering, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| | - Dennis J. Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio-44196, USA
| |
Collapse
|
40
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [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] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
41
|
Chen Y, Yuan Y, Peng L, Dong X, Xu Y, Wang Y, Yang Y. Effects of increasing sensitizing doses of ovalbumin on airway hyperresponsiveness in asthmatic mice. Immun Inflamm Dis 2024; 12:e1225. [PMID: 38533918 PMCID: PMC10966913 DOI: 10.1002/iid3.1225] [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: 10/27/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND The dosage of ovalbumin (OVA) during the sensitization stage is considered a crucial factor in the development of airway hyperresponsiveness (AHR). However, the inconsistent dosages of sensitizing OVA used in current studies and the lack of research on their impact on AHR are notable limitations. METHODS We examined the impact of increasing sensitizing doses of OVA in a murine asthma model, which entailed initial sensitization with OVA followed by repeated exposure to OVA aerosols. BALB/c mice were primed with doses of OVA (0, 10, 20, 50, and 100 μg) plus 1 mg Alum on Days 0 and 7, and were challenged with OVA aerosols (10 mg/mL for 30 min) between Days 14 and 17. Antigen-induced AHR to methacholine (MCh), as well as histological changes, eosinophilic infiltration, and epithelial injury were assessed. RESULTS The result indicated that there are striking OVA dose-related differences in antigen-induced AHR to MCh. The most intense antigen-induced AHR to MCh was observed with sensitization at 50 μg, while weaker responses were seen at 10, 20, and 100 μg. Meanwhile, there was a significant increase in eosinophil count with sensitization at 50 μg. The changes of AHR were correlated with total cells count, lymphocytes count, eosinophils count, and basophils count in bronchoalveolar lavage fluid; however, it did not correlate with histological changes such as cellular infiltration into bronchovascular bundles and goblet cell hyperplasia of the bronchial epithelium. CONCLUSION Overall, this study demonstrated that sensitization with 50 μg of OVA resulted in the most significant AHR compared to other dosages. These findings may offer valuable insights for future research on mouse asthma modeling protocols.
Collapse
Affiliation(s)
- Yan‐Jiao Chen
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and MeridianYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghaiP.R. China
| | - Yu Yuan
- Deparment of Acupuncture and MoxibustionGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiP.R. China
| | - Lu Peng
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and MeridianYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghaiP.R. China
| | - Xin‐Yi Dong
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and MeridianYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghaiP.R. China
| | - Yu‐Dong Xu
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and MeridianYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghaiP.R. China
| | - Yu Wang
- Shanghai University of Traditional Chinese MedicineShanghaiP.R. China
| | - Yong‐Qing Yang
- Shanghai University of Traditional Chinese MedicineShanghaiP.R. China
| |
Collapse
|
42
|
Fröhlich E. Animals in Respiratory Research. Int J Mol Sci 2024; 25:2903. [PMID: 38474149 DOI: 10.3390/ijms25052903] [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: 01/17/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The respiratory barrier, a thin epithelial barrier that separates the interior of the human body from the environment, is easily damaged by toxicants, and chronic respiratory diseases are common. It also allows the permeation of drugs for topical treatment. Animal experimentation is used to train medical technicians, evaluate toxicants, and develop inhaled formulations. Species differences in the architecture of the respiratory tract explain why some species are better at predicting human toxicity than others. Some species are useful as disease models. This review describes the anatomical differences between the human and mammalian lungs and lists the characteristics of currently used mammalian models for the most relevant chronic respiratory diseases (asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary hypertension, pulmonary fibrosis, and tuberculosis). The generation of animal models is not easy because they do not develop these diseases spontaneously. Mouse models are common, but other species are more appropriate for some diseases. Zebrafish and fruit flies can help study immunological aspects. It is expected that combinations of in silico, in vitro, and in vivo (mammalian and invertebrate) models will be used in the future for drug development.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
43
|
Kazim M, Razian SA, Zamani E, Varandani D, Shahbad R, Desyatova A, Jadidi M. Variability in structure, morphology, and mechanical properties of the descending thoracic and infrarenal aorta around their circumference. J Mech Behav Biomed Mater 2024; 150:106332. [PMID: 38160644 DOI: 10.1016/j.jmbbm.2023.106332] [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: 10/27/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Aortic diseases, such as aneurysms, atherosclerosis, and dissections, demonstrate a preferential development and progression around the aortic circumference, resulting in a highly heterogeneous disease state around the circumference. Differences in the aorta's structural composition and mechanical properties may be partly responsible for this phenomenon. Our goal in this study was to analyze the mechanical and structural properties of the human aorta at its lateral, anterior, posterior, and medial quadrants in two regions prone to circumferentially inhomogeneous diseases, descending Thoracic Aorta (TA) and Infrarenal Aorta (IFR). Human aortas were obtained from 10 donors (64 ± 11 years) and dissected from their loose surrounding tissue. Mechanical properties were determined in all four quadrants of TA and IFR using planar biaxial testing and fitted to three common constitutive models. The structure of tissues was assessed using Movat Pentachrome stained histology slides. We observed that the anterior quadrant exhibited the greatest thickness, followed by the lateral region, in both the TA and IFR. In TA, the posterior wall appeared as the stiffest location in most samples, while in IFR, the anterior wall was the stiffest. We observed a higher glycosaminoglycans content in the lateral and posterior regions of the IFR. We found elastin density to be similar in TA lateral, anterior, and posterior quadrants, while in IFR, the anterior region demonstrated the highest elastin density. Despite significant variations between subjects, this study highlights the distinct morphometrical, mechanical, and structural properties between the quadrants of both TA and IFR.
Collapse
Affiliation(s)
- Madihah Kazim
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | | | - Elham Zamani
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | - Dheeraj Varandani
- Department of Computer Science, University of Nebraska Omaha, Omaha, NE, USA
| | - Ramin Shahbad
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | | | - Majid Jadidi
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA.
| |
Collapse
|
44
|
Zhang Y, Liang Z, Xing H, Yu C, Liang J, Xu Q, Song J, He Z. A model of pregnancy-associated malaria for inducing adverse pregnancy outcomes in ICR mouse. Exp Parasitol 2024; 257:108686. [PMID: 38158008 DOI: 10.1016/j.exppara.2023.108686] [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: 06/08/2023] [Revised: 11/08/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Based on understanding of placental pathological features and safe medication in pregnancy-associated malaria (PAM), establishment of a stable pregnant mouse infection model with Plasmodium was urgently needed. METHODS ICR mice with vaginal plugs detected were randomly divided into post-pregnancy infection (Malaria+) and uninfected pregnancy (Malaria-) cohorts. Age-matched mice that had not been mated were infected as pre-pregnancy infection group (Virgin control), which were subsequently mated with ICR males. All mice were inoculated with 1 × 106Plasmodium berghei ANKA-infected RBCs by intraperitoneal injection, and the same amount of saline was given to Malaria- group. We recorded the incidence of adverse pregnancy outcomes and the amounts of offspring in each group. RESULTS The Virgin group mice were unable to conceive normally, and vaginal bleeding, abortion, or stillbirth appeared in the Malaria+ group. The incidence of adverse pregnancy outcomes was extremely high and statistically significant compared with the control (Malaria-) group (P < 0.05), of which placenta exhibited pathological features associated with human gestational malaria. CONCLUSIONS The intraperitoneal injection of 1 × 106Plasmodium berghei ANKA-infected RBCs could establish a model of pregnancy-associated malaria in ICR mouse.
Collapse
Affiliation(s)
- Yingying Zhang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhiming Liang
- Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Haoyu Xing
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chuyi Yu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianming Liang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhouqing He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
45
|
Neary MT, Mulder LM, Kowalski PS, MacLoughlin R, Crean AM, Ryan KB. Nebulised delivery of RNA formulations to the lungs: From aerosol to cytosol. J Control Release 2024; 366:812-833. [PMID: 38101753 DOI: 10.1016/j.jconrel.2023.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
In the past decade RNA-based therapies such as small interfering RNA (siRNA) and messenger RNA (mRNA) have emerged as new and ground-breaking therapeutic agents for the treatment and prevention of many conditions from viral infection to cancer. Most clinically approved RNA therapies are parenterally administered which impacts patient compliance and adds to healthcare costs. Pulmonary administration via inhalation is a non-invasive means to deliver RNA and offers an attractive alternative to injection. Nebulisation is a particularly appealing method due to the capacity to deliver large RNA doses during tidal breathing. In this review, we discuss the unique physiological barriers presented by the lung to efficient nebulised RNA delivery and approaches adopted to circumvent this problem. Additionally, the different types of nebulisers are evaluated from the perspective of their suitability for RNA delivery. Furthermore, we discuss recent preclinical studies involving nebulisation of RNA and analysis in in vitro and in vivo settings. Several studies have also demonstrated the importance of an effective delivery vector in RNA nebulisation therefore we assess the variety of lipid, polymeric and hybrid-based delivery systems utilised to date. We also consider the outlook for nebulised RNA medicinal products and the hurdles which must be overcome for successful clinical translation. In summary, nebulised RNA delivery has demonstrated promising potential for the treatment of several lung-related conditions such as asthma, COPD and cystic fibrosis, to which the mode of delivery is of crucial importance for clinical success.
Collapse
Affiliation(s)
- Michael T Neary
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | | | - Piotr S Kowalski
- School of Pharmacy, University College Cork, Ireland; APC Microbiome, University College Cork, Cork, Ireland
| | | | - Abina M Crean
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | - Katie B Ryan
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland.
| |
Collapse
|
46
|
Yılmaz D, Mathavan N, Wehrle E, Kuhn GA, Müller R. Mouse models of accelerated aging in musculoskeletal research for assessing frailty, sarcopenia, and osteoporosis - A review. Ageing Res Rev 2024; 93:102118. [PMID: 37935249 DOI: 10.1016/j.arr.2023.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Musculoskeletal aging encompasses the decline in bone and muscle function, leading to conditions such as frailty, osteoporosis, and sarcopenia. Unraveling the underlying molecular mechanisms and developing effective treatments are crucial for improving the quality of life for those affected. In this context, accelerated aging models offer valuable insights into these conditions by displaying the hallmarks of human aging. Herein, this review focuses on relevant mouse models of musculoskeletal aging with particular emphasis on frailty, osteoporosis, and sarcopenia. Among the discussed models, PolgA mice in particular exhibit hallmarks of musculoskeletal aging, presenting early-onset frailty, as well as reduced bone and muscle mass that closely resemble human musculoskeletal aging. Ultimately, findings from these models hold promise for advancing interventions targeted at age-related musculoskeletal disorders, effectively addressing the challenges posed by musculoskeletal aging and associated conditions in humans.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland; AO Research Institute Davos, Davos Platz, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
47
|
Diwan R, Bhatt HN, Beaven E, Nurunnabi M. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204:115147. [PMID: 38065244 PMCID: PMC10787600 DOI: 10.1016/j.addr.2023.115147] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, and life-threatening interstitial lung disease which causes scarring in the lung parenchyma and thereby affects architecture and functioning of lung. It is an irreversible damage to lung functioning which is related to epithelial cell injury, immense accumulation of immune cells and inflammatory cytokines, and irregular recruitment of extracellular matrix. The inflammatory cytokines trigger the differentiation of fibroblasts into activated fibroblasts, also known as myofibroblasts, which further increase the production and deposition of collagen at the injury sites in the lung. Despite the significant morbidity and mortality associated with PF, there is no available treatment that efficiently and effectively treats the disease by reversing their underlying pathologies. In recent years, many therapeutic regimens, for instance, rho kinase inhibitors, Smad signaling pathway inhibitors, p38, BCL-xL/ BCL-2 and JNK pathway inhibitors, have been found to be potent and effective in treating PF, in preclinical stages. However, due to non-selectivity and non-specificity, the therapeutic molecules also result in toxicity mediated severe side effects. Hence, this review demonstrates recent advances on PF pathology, mechanism and targets related to PF, development of various drug delivery systems based on small molecules, RNAs, oligonucleotides, peptides, antibodies, exosomes, and stem cells for the treatment of PF and the progress of various therapeutic treatments in clinical trials to advance PF treatment.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States; The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
48
|
Smith J, Richerson G, Kouchi H, Duprat F, Mantegazza M, Bezin L, Rheims S. Are we there yet? A critical evaluation of sudden and unexpected death in epilepsy models. Epilepsia 2024; 65:9-25. [PMID: 37914406 DOI: 10.1111/epi.17812] [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: 08/04/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/03/2023]
Abstract
Although animal models have helped to elaborate meaningful hypotheses about the pathophysiology of sudden and unexpected death in epilepsy (SUDEP), specific prevention strategies are still lacking, potentially reflecting the limitations of these models and the intrinsic difficulties of investigating SUDEP. The interpretation of preclinical data and their translation to diagnostic and therapeutic developments in patients thus require a high level of confidence in their relevance to model the human situation. Preclinical models of SUDEP are heterogeneous and include rodent and nonrodent species. A critical aspect is whether the animals have isolated seizures exclusively induced by a specific trigger, such as models where seizures are elicited by electrical stimulation, pharmacological intervention, or DBA mouse strains, or whether they suffer from epilepsy with spontaneous seizures, with or without spontaneous SUDEP, either of nongenetic epilepsy etiology or from genetically based developmental and epileptic encephalopathies. All these models have advantages and potential disadvantages, but it is important to be aware of these limitations to interpret data appropriately in a translational perspective. The majority of models with spontaneous seizures are of a genetic basis, whereas SUDEP cases with a genetic basis represent only a small proportion of the total number. In almost all models, cardiorespiratory arrest occurs during the course of the seizure, contrary to that in patients observed at the time of death, potentially raising the issue of whether we are studying models of SUDEP or models of periseizure death. However, some of these limitations are impossible to avoid and can in part be dependent on specific features of SUDEP, which may be difficult to model. Several preclinical tools are available to address certain gaps in SUDEP pathophysiology, which can be used to further validate current preclinical models.
Collapse
Affiliation(s)
- Jonathon Smith
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - George Richerson
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Hayet Kouchi
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - Fabrice Duprat
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| | - Massimo Mantegazza
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| | - Laurent Bezin
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - Sylvain Rheims
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and Lyon 1 University, Lyon, France
| |
Collapse
|
49
|
Hemström P, Jugg B, Watkins R, Jonasson S, Elfsmark L, Rutter S, Åstot C, Lindén P. Phospholipid chlorohydrins as chlorine exposure biomarkers in a large animal model. Toxicol Lett 2024; 391:32-38. [PMID: 38048885 DOI: 10.1016/j.toxlet.2023.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
Chlorine is a toxic industrial chemical that has been used as a chemical weapon in recent armed conflicts. Confirming human exposure to chlorine has proven challenging, and there is currently no established method for analyzing human biomedical samples to unambiguously verify chlorine exposure. In this study, two chlorine-specific biomarkers: palmitoyl-oleoyl phosphatidylglycerol chlorohydrin (POPG-HOCl) and the lipid derivative oleoyl ethanolamide chlorohydrin (OEA-HOCl) are shown in bronchoalveolar lavage fluid (BALF) samples from spontaneously breathing pigs after chlorine exposure. These biomarkers are formed by the chemical reaction of chlorine with unsaturated phospholipids found in the pulmonary surfactant, which is present at the gas-liquid interface within the lung alveoli. Our results strongly suggest that lipid chlorohydrins are promising candidate biomarkers in the development of a verification method for chlorine exposure. The establishment of verified methods capable of confirming the illicit use of toxic industrial chemicals is crucial for upholding the principles of the Chemical Weapons Convention (CWC) and enforcing the ban on chemical weapons. This study represents the first published dataset in BALF revealing chlorine biomarkers detected in a large animal. Furthermore, these biomarkers are distinct in that they originate from molecular chlorine rather than hypochlorous acid.
Collapse
Affiliation(s)
- Petrus Hemström
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | | | | | - Sofia Jonasson
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | - Linda Elfsmark
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | | | - Crister Åstot
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | - Pernilla Lindén
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden.
| |
Collapse
|
50
|
Hapuarachi B, Danson S, Wadsley J, Muthana M. Exercise to transform tumours from cold to hot and improve immunotherapy responsiveness. Front Immunol 2023; 14:1335256. [PMID: 38149260 PMCID: PMC10749948 DOI: 10.3389/fimmu.2023.1335256] [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: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Exercise provides significant health benefits to patients diagnosed with cancer including improved survival outcomes, quality of life and reduced cancer recurrence. Across multiple murine cancer models, aerobic exercise and resistance training has exhibited anti-tumour properties illustrated by inhibited tumour growth, reduced metastatic potential and modulation of the tumour microenvironment to allow the recognition and destruction of cancer cells. Clinical studies have demonstrated the rapid mobilisation and circulatory release of mature lymphoid populations, myokines and cytokines that occurs with exercise along with tumour vasculature normalisation. Tumour microenvironments enriched with immune cells with anti-cancer potential, such as CD8+ T cells, are termed 'hot', whilst those favouring an immunosuppressive environment and lacking in effector immune cells are classed as 'cold'. Pre-clinical evidence suggests exercise training has the potential to reprogramme cold tumours to become hot, although this requires validation in clinical studies. This hot environment could potentiate immunotherapy responsiveness, improving survival outcomes of patients undergoing cancer immunotherapy and allow those with typically cold tumours to benefit from immunotherapy. This review discusses the complex interactions between exercise and cancer, including exercise-induced alterations within the tumour microenvironment and systemic immunity. The potential role exercise may play in improving cancer immunotherapy responsiveness is explored. This review also highlights the need for translational studies exploring the role of exercise in patients with cancer with the potential to widen the spectrum of tumours that derive significant benefit from immunotherapy.
Collapse
Affiliation(s)
- Brindley Hapuarachi
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Sarah Danson
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Jon Wadsley
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Munitta Muthana
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
| |
Collapse
|