1
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|
2
|
Jenkins BW, Moore CF, Jantzie LL, Weerts EM. Prenatal cannabinoid exposure and the developing brain: Evidence of lasting consequences in preclinical rodent models. Neurosci Biobehav Rev 2025; 175:106207. [PMID: 40373945 DOI: 10.1016/j.neubiorev.2025.106207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
Cannabis use by people who are pregnant is increasing. Understanding how prenatal cannabinoid exposure (PCE) affects infants and children is of high public health significance. Epidemiological studies have associated PCE with cognitive symptoms, including impaired learning, memory, attention, and executive control, and affective symptoms, including anxiety, emotional dysregulation, and social impairments, in children, adolescents, and young adults. PCE is also associated with neurobiological changes including decreased corticolimbic white matter and functional connectivity; however, the underlying mechanisms for these persisting effects remain unknown. Rodent models are essential for uncovering the effects of PCE on the developing brain. This review summarizes rodent studies focused on the cognitive and affective behavioral and neurobiological outcomes of PCE. Rodent studies have reported cognitive deficits, including impaired learning, memory, attention, and executive control, and affect-related impairments, including anxiety-like behavior, altered stress coping, social impairments, and anhedonia-like behavior, in adolescent and adult offspring. Studies have also demonstrated that PCE affects several underlying neurotransmitter systems, producing dopamine hyperactivity, glutamate and serotonin hypoactivity, and dysregulating GABA and opioid signaling. Evidence further suggests a marked difference in outcomes between males and females, with males being more susceptible to the enduring effects of PCE. However, studies that investigate female-specific outcomes or sex as a biological variable are scarce. Altogether, rodent studies provide corroborating evidence that PCE produces lasting cognitive and affective impairments underpinned by altered neurobiological mechanisms. Research is critically needed to improve our understanding of the risks associated with cannabis use during pregnancy and effects across the lifespan.
Collapse
Affiliation(s)
- Bryan W Jenkins
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA
| | - Catherine F Moore
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA
| | - Lauren L Jantzie
- Departments of Pediatrics, Neurosurgery, and Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Elise M Weerts
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA.
| |
Collapse
|
3
|
de Miguel R, Vallejo R, Kegler K, Kreutzer R, Mayoral FJ, Okazaki Y, Ortega P, Polledo L, Razinger T, Richard OK, Sanchez R, Warfving N, Domènech A, Weber K. Onset and progression of postmortem histological changes in the kidneys of RccHan TM:WIST rats. Front Vet Sci 2025; 12:1578579. [PMID: 40417372 PMCID: PMC12100932 DOI: 10.3389/fvets.2025.1578579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/31/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Death initiates a cascade of physiological and biochemical alterations in organs and tissues, resulting in microscopic changes that challenge the histopathological evaluation. The aim of this study was to compile and illustrate the microscopic changes in the kidneys of rats subjected to delayed postmortem fixation. It also scrutinizes the influence of exsanguination, cooling methods and air circulation on the initiation and progression of these alterations. Methods Twenty-four Wistar Han outbred rats (RccHanTM:WIST) were sacrificed and stored either at room temperature (18-22°C; half of the carcasses were exsanguinated after sacrifice) or under refrigeration (2-4°C). Necropsies were conducted at different time points postmortem (i.e., 0.5, 1, 4, 8, 12, 24, 36, 48 h for carcasses stored at room temperature, and 7 and 14 days for carcasses stored under refrigeration). Kidney sections underwent simultaneous digital evaluation by 14 pathologists until a consensus was reached on the key findings, terminology, and intensity levels. Results When stored at room temperature, the first changes were seen after 4 h, and involved distal convoluted tubules and inner stripe of the outer medulla. After 8 h, all structures except glomeruli were affected. Alterations were similar in quality and intensity after 36 h as after 48 h. Exsanguination delayed the onset of postmortem changes and slightly decreased their overall severity at any given timepoint. The nature of the changes under refrigeration was like those alterations noted in animals stored at room temperature. The intensity of postmortem changes observed after 7 and 14 days under refrigeration was similar to those recorded after 48 h at room temperature. No clear differences were observed between animals stored in a closed plastic bag and animals stored in a perforated cardboard box to allow air circulation. Conclusion This work elucidates the onset and progression of postmortem changes in the kidneys of Wistar Han rats, offering insights to accurately differentiate them from real changes and enhance histopathological evaluation.
Collapse
|
4
|
Kelley J, Li H, Sun Y, Ren P, Chen G, Sun S, Zhao J, Buchweitz N, Kern M, Reitman CA, Townsend DM, Yao H, Wu Y. Endplate remodeling: a key indicator of cigarette smoke exposure-induced intervertebral disc degeneration in a male rat model. JBMR Plus 2025; 9:ziaf016. [PMID: 40176949 PMCID: PMC11961068 DOI: 10.1093/jbmrpl/ziaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 04/05/2025] Open
Abstract
Recent clinical studies have established a strong association between cigarette smoking and degenerative disc disease. Both in vitro and in vivo research indicated that cigarette smoke disrupts cellular homeostasis in the intervertebral disc (IVD), leading to spatiotemporal remodeling of the extracellular matrix, with a notable reduction in solute diffusivity within the cartilage endplate (CEP). As the CEP serves as a critical mechanical barrier and solute diffusion pathway for the IVD, both roles can be compromised by pathological changes in the tissue. This underscores the need for a more comprehensive examination of endplate remodeling during IVD degeneration, particularly in the context of cigarette smoking and cessation. The objective of this study was to perform a quantitative analysis of the structure-material property relationship changes in the endplate at tissue and cellular levels to determine how endplate mineralization progresses during IVD degeneration in the context of cigarette smoke exposure and cessation, using our previously developed Sprague-Dawley rat model. Our results indicate that cigarette smoke exposure-induced endplate remodeling is characterized by a higher CEP histological grade, increased aberrant CEP calcification level, and elevated bony endplate surface flatness score, all of which correlated with an accelerated chondrocyte cell life cycle. Smoke cessation alone was insufficient to reverse the mineralization progression in the endplate. Principal component analysis further identified alterations in endplate morphometry at the tissue level and disruptions in the chondrocyte life cycle at cellular level as key markers of degenerative remodeling. These findings establish endplate remodeling as a key indicator of smoke exposure-induced IVD degeneration and inform the development of novel therapeutic strategies aimed at preserving or improving disc health.
Collapse
Affiliation(s)
- Joshua Kelley
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
| | - Hui Li
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
| | - Yi Sun
- Department of Orthopedics, The 2nd Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Pengling Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, 100050 Beijing, China
| | - Guanghua Chen
- Department of Orthopedics, The 2nd Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Shuchun Sun
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
| | - Jichao Zhao
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
| | - Nathan Buchweitz
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
| | - Michael Kern
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, 29425 Charleston, SC, United States
| | - Charles A Reitman
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, 29425 Charleston, SC, United States
| | - Danyelle M Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 29425 Charleston, SC, United States
| | - Hai Yao
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
- Department of Oral Health Sciences, Medical University of South Carolina, 29425 Charleston, SC, United States
| | - Yongren Wu
- Department of Bioengineering, Clemson University, 29425 Charleston, SC, United States
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, 29425 Charleston, SC, United States
| |
Collapse
|
5
|
Wanner IB, McCabe JT, Huie JR, Harris NG, Paydar A, McMann-Chapman C, Tobar A, Korotcov A, Burns MP, Koehler RC, Wan J, Allende Labastida J, Tong J, Zhou J, Davis LM, Radabaugh HL, Ferguson AR, Van Meter TE, Febo M, Bose P, Wang KK, Kobeissy F, Apiliogullari S, Zhu J, Rubenstein R, Awwad HO. Prospective Harmonization, Common Data Elements, and Sharing Strategies for Multicenter Pre-Clinical Traumatic Brain Injury Research in the Translational Outcomes Project in Neurotrauma Consortium. J Neurotrauma 2025; 42:877-897. [PMID: 39831841 DOI: 10.1089/neu.2023.0653] [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] [Indexed: 01/22/2025] Open
Abstract
Effective team science requires procedural harmonization for rigor and reproducibility. Multicenter studies across experimental modalities (domains) can help accelerate translation. The Translational Outcomes Project in NeuroTrauma (TOP-NT) is a pre-clinical traumatic brain injury (TBI) consortium charged with establishing and validating noninvasive TBI assessment tools through team science. Here, we present practical approaches for harmonization of TBI research across five centers providing needed vocabulary and structure to achieve centralized data organization and use. This includes data sharing as an essential step that enables validating data between domains, evaluating reproducibility between sites, and performing multimodal analyses. As part of this process, TOP-NT (1) produced a library of TBI-relevant standard operating procedures to coordinate workflow, (2) aligned 481 pre-clinical and clinical common data elements (CDEs), and (3) generated 272 new pre-clinical TBI CDEs. This consortium then (4) connected diverse data types to validate assessments across domains and to allow multivariable TBI phenotyping. Lastly, TOP-NT (5) specified technical quality controls for pre-clinical studies. These harmonization tools can facilitate reproducibility in team science, help distinguish a wide injury spectrum from technical variability, apply quality-controls, and ease higher level data analyses. TOP-NT uses three rat TBI models across four sites. Each site collects primary outcome measures, including magnetic resonance imaging (MRI) protocols and blood biomarkers of neuronal and glial injury, validated by histopathology and behavioral outcomes. Collected data are organized using the 481 TOP-NT pre-clinical CDEs, covering surgical, behavioral, biomarker, MRI, and quantitative histopathological methods. We report data curation steps suited for data storage using the Open Data Commons for TBI as a centralized data repository, allowing unbiased cross-site analysis. This approach leads to introducing a higher level, syndromic understanding of TBI signatures. TOP-NT authors outline a semantic and structural framework suggesting strategies for robust pre-clinical research in multicenter trials to improve translatability for TBI assessments. [Figure: see text].
Collapse
Affiliation(s)
- Ina-Beate Wanner
- Intellectual and Developmental Disability Center (IDDRC), David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, California, USA
| | - Joseph T McCabe
- Department of Anatomy, Physiology & Genetics, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - J Russell Huie
- Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, California, USA
- Principal Investigator, Veterans Affairs Healthcare System, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Neil G Harris
- Department of Neurosurgery, Brain Research Injury Center (BIRC), Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Afshin Paydar
- Department of Neurosurgery, Brain Research Injury Center (BIRC), Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Chloe McMann-Chapman
- Intellectual and Developmental Disability Center (IDDRC), David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, California, USA
| | - Anthony Tobar
- Semel Institute for Neuroscience and Human Behavior, IDDRC, Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Alexandru Korotcov
- Department of Radiology & Radiological Sciences, Uniformed Services University, Bethesda, Maryland, USA
| | - Mark P Burns
- Georgetown University Medical Center, Center for Neural Injury and Repair, Washington, District of Columbia, USA
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Javier Allende Labastida
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan Tong
- Semel Institute for Neuroscience and Human Behavior, IDDRC, Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Jinyuan Zhou
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lex Maliga Davis
- Brain and Spinal Injury Center, University of California, San Francisco, California, USA
| | - Hannah L Radabaugh
- Brain and Spinal Injury Center, University of California, San Francisco, California, USA
| | - Adam R Ferguson
- Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, California, USA
- Principal Investigator, Veterans Affairs Healthcare System, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | | | - Marcelo Febo
- Departmet Psychiatry, University of Florida, Gainesville, Florida, USA
- Department of Psychiatry, Advanced Magnetic Resonance Imaging and Spectroscopy Facility, University of Florida, Gainesville, Florida, USA
- Department of Psychiatry (Room L4-100F), McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Prodip Bose
- Department of Anesthesiology, and Department of Neurology at the College of Medicine, University of Florida, Gainesville, Florida, USA
- Brain Rehabilitation Research (Center), Malcom Randall VAMC, Gainesville, Florida, USA
| | - Kevin K Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Seza Apiliogullari
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Jiepei Zhu
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Richard Rubenstein
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Hibah O Awwad
- Division of Neuroscience, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
6
|
Massey N, Vasanthi SS, Gimenez-Lirola LG, Tyler H, Thippeswamy T. Proinflammatory cytokines, oxidative stress, and organ function as biomarkers of soman (GD) chronic neurotoxicity. Sci Rep 2025; 15:9021. [PMID: 40089647 PMCID: PMC11910520 DOI: 10.1038/s41598-025-94190-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Organophosphate (OP) nerve agents, such as soman (GD), pose great risk to neurological health by inhibiting acetylcholinesterase, leading to seizures, epilepsy, and behavioral deficits. While acute treatment may alleviate immediate symptoms, the long-term consequences, particularly those involving neuroinflammation and systemic toxicity, remain poorly understood. This study used adult male and female Sprague Dawley rats to investigate the chronic effects of a single acute exposure to soman (132 µg/kg, s.c., 1.2 × LD50) on neuroinflammation, behavioral comorbidity, and systemic toxicity. Following exposure, animals were treated with atropine sulfate (2 mg/kg, i.m.) and oxime HI-6 (125 mg/kg, i.m.) to mitigate peripheral cholinergic effects, and with midazolam (3 mg/kg, i.m., 1 h post-exposure) to control seizures. Spontaneously recurring seizures were monitored during handling and with video electroencephalogram (vEEG). Neurobehavioral deficits were assessed 4-8 weeks post-exposure. At 18 weeks post-exposure, brain, serum, and cerebrospinal fluid (CSF) were analyzed for inflammatory and nitro-oxidative stress markers, and the liver and kidney function biomarkers were evaluated. Soman-exposed animals developed epilepsy, confirmed by handling-induced seizures and/or continuous vEEG monitoring. Behavioral assessments revealed significant memory deficits following soman exposure. Proinflammatory cytokines (TNF-α, IL-6, IL-1α, IL-18, IL-17A, and MCP-1) were significantly elevated in both serum and CSF, alongside corresponding increases in their gene expression in the brain. Elevated reactive oxygen/nitrogen species were detected in the serum. Although hematological parameters remained unchanged, a significant increase in total bilirubin and an upward trend in serum blood urea nitrogen (BUN) levels and BUN: Creatinine ratio indicated potential liver and kidney dysfunction. However, no significant structural changes in these organs at the cellular level were observed in histological analyses. This study identifies critical chronic biomarkers of soman exposure affecting the brain, serum, CSF, liver, and kidney. The findings highlight the critical need to monitor systemic and neurological impacts, as well as organ function, to develop effective diagnostic and therapeutic strategies for survivors of nerve agent exposure or OP pesticide poisoning. Behavioral deficits and EEG changes in soman-exposed animals further emphasize the long-term neurological consequences of exposure.
Collapse
Affiliation(s)
- Nyzil Massey
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Suraj S Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Luis G Gimenez-Lirola
- Vet Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Harm Tyler
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
7
|
Scott KJ, Bilkey DK. Sex-dependent effects of rat maternal immune activation on motor function in offspring of poly I:C treated rats. Behav Brain Res 2025; 481:115431. [PMID: 39814236 DOI: 10.1016/j.bbr.2025.115431] [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: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
A majority of people with schizophrenia will experience motor symptoms such as impairments to coordination, balance and motor sequencing. These neurological soft signs are associated with negative social and functional outcomes, and poor disease prognosis. They occur prior to medication exposure, suggesting they are an intrinsic feature of schizophrenia. Despite the need to better understand this dysfunction, relatively few studies have provided a detailed focus on motor capability in animal models of schizophrenia. Here we investigate motor coordination in a rat maternal immune activation (MIA) model of schizophrenia risk. The female and male offspring of Polyinosinic:polycytidylic acid (Poly I:C), and vehicle-treated, pregnant dams were tested in a horizontal ladder rung task using regular and irregular rung configurations. We extracted information about limb positions from video, and measured faults and gait coordination in the task. We found that adult male MIA rats were more likely to slip from the ladder rungs than control animals, and they were more likely to have multiple limbs slip simultaneously. MIA rats also exhibited more variability in stride length, a result that correlated with slips and mirrored disease-related changes in human gait. In contrast, female MIA rats displayed minimal alterations in motor performance. Our findings show that the ladder task uncovers sex-dependent effects on motor coordination in MIA rats and highlights the potential usefulness of the MIA model for investigating motor dysfunction in an animal model of schizophrenia risk.
Collapse
Affiliation(s)
- K Jack Scott
- Department of Psychology, University of Otago, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, New Zealand.
| |
Collapse
|
8
|
Tulva K, Pirajev A, Zeb A, Aksoy AE, Bello A, Lee B, Guðjónsson BF, Helgadottir SB, Jagomäe T, García-Llorca A, Eysteinsson T, Jürgenson M, Plaas M, Vasar E, Kaasik A, Hickey MA. Early trigeminal and sensory impairment and lysosomal dysfunction in accurate models of Wolfram syndrome. Exp Neurol 2025; 385:115099. [PMID: 39662795 DOI: 10.1016/j.expneurol.2024.115099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/04/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Wolfram syndrome (WS) is a rare condition caused by homozygous or compound heterozygous mutations in the WFS1 gene primarily. It is diagnosed on the basis of early-onset diabetes mellitus and optic nerve atrophy. Patients complain of trigeminal-like migraines and show deficits in vibration sensation, but the underlying cause is unknown. Using accurate cell models and two separate, accurate rodent models of WS that show excellent face and construct validity, here we have examined trigeminus, sensation and sensory neuronal function in WS. Analysis of ex vivo and in vivo MRI sequences revealed profound trigeminal atrophy in each rodent model, a novel finding in WS. Optic nerve atrophy is a diagnostic sign in WS, and trigeminal atrophy occurred at the time of earliest loss of optic nerve volume. We also observed deficits in mechanical sensation in our mouse WS model, and pathological analysis revealed extensive inflammation in trigeminal sensory nucleus, both of which are novel findings in WS. Sensory neurons (dorsal root ganglia) showed impaired calcium handling upon depolarisation and reduced mitochondrial membrane potential. Finally, lysosomes were smaller, soma lysosome content was decreased and importantly, lysosome acidity was impaired in sensory neurons, all of which are novel findings in WS. We validated these findings using two separate publicly available datasets, both from WS patient fibroblast-derived neural stem cells. We observed a highly significant functional enrichment of GO cellular component lysosome-related terms among the differentially expressed proteins and genes, with the majority of lysosome-related proteins being downregulated. These data reveal extensive impairments in the trigeminal pathway and nociceptive neurons in WS that may contribute to trigeminal and sensory symptoms observed in patients. Moreover, we note that mutations in WFS1 are relatively common and, given the importance of WFS1 for sensory function, our data may also shed light on sensory impairments in general.
Collapse
Affiliation(s)
- Kerli Tulva
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Aleksander Pirajev
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Akbar Zeb
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Asya E Aksoy
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Azizah Bello
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Benjamin Lee
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Baldvin F Guðjónsson
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Sigridur B Helgadottir
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Toomas Jagomäe
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411 Tartu, Estonia; Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Andrea García-Llorca
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | - Thor Eysteinsson
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | - Monika Jürgenson
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411 Tartu, Estonia; Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Allen Kaasik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | - Miriam A Hickey
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411 Tartu, Estonia.
| |
Collapse
|
9
|
Varela MF, Oria M, Poling HM, Lopriore E, Peiro JL. Development and Validation of a Minimally Invasive Transuterine Experimental Model of Gastroschisis. J Pediatr Surg 2025; 60:162163. [PMID: 39823694 DOI: 10.1016/j.jpedsurg.2025.162163] [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: 10/05/2024] [Revised: 12/13/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Perinatal management of gastroschisis remains a subject of substantial research. Current models, including teratogenic, genetic, and surgical approaches, often fail to accurately replicate gastroschisis, exhibiting limitations such as inaccurate phenotyping, low success rates, high mortality, lack of scientific validation, and significant technical challenges. Refined disease models are essential for improving the understanding of GS. This study seeks to develop and validate a minimally invasive transuterine experimental model of GS that overcomes these existing constraints to advance gastroschisis research. METHODS A gastroschisis model was surgically created in rat fetuses at E17 (n = 51 fetuses from n = 13 dams). Intestines were harvested at term and divided into herniated gastroschisis (GS-H), intra-abdominal gastroschisis (GS-I), and control (Co) groups. Morphometric analysis, histopathological examination, immunohistochemistry for interstitial cells of Cajal (ICC), double immunofluorescence for ICC and mast cells, TUNEL assay for apoptotic cells, and multiplex cytokine assay were performed to assess intestinal architecture, inflammation, ICC network, apoptosis, and cytokine levels across studied groups. RESULTS Histology from GS intestines revealed subchronic inflammation, peel formation, and architectural disruption. Herniated intestines exhibited a significantly increased weight/length ratio and thicker outer layers (p < 0.001) compared with control intestines. Herniated intestines had elevated inflammatory cytokine levels (GS-H vs GS-I and Co, p < 0.05 for G-CSF, GM-CSF, IL-12p70, IL-1beta) and increased apoptotic activity. CONCLUSIONS We developed and validated a new surgical model of GS that offers improved survival and feasibility. The key morphological changes and molecular markers observed in this experimental model resemble human gastroschisis.
Collapse
Affiliation(s)
- Maria Florencia Varela
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Neonatology, Department of Pediatrics, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Marc Oria
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Holly Marie Poling
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Jose Luis Peiro
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA.
| |
Collapse
|
10
|
Ciesielski V, Guerbette T, Fret L, Succar M, Launay Y, Dahirel P, Legrand P, Vlach M, Blat S, Rioux V. Dietary pentadecanoic acid supplementation at weaning in essential fatty acid-deficient rats shed light on the new family of odd-chain n-8 PUFAs. J Nutr Biochem 2025; 137:109814. [PMID: 39617355 DOI: 10.1016/j.jnutbio.2024.109814] [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: 05/10/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/22/2024]
Abstract
Pentadecanoic acid (C15:0) is a saturated odd-chain fatty acid (OCFA), mainly found in dairy products. Its physiological and nutritional effects are still unknown, yet some recent evidences suggest it might be beneficial to human health. Moreover, pentadecanoic acid has recently been suspected of having essential roles in humans, although the mechanisms are not described. We therefore questioned the potential essentiality of this fatty acid (FA). We investigated in vivo the effect of a C15:0 supplementation on essential fatty acid (EFA) deficient Wistar rats. Female rats were fed an EFA-deficient diet 2 weeks before mating, during pregnancy and lactation. Weaned pups were fed the EFA-deficient diet or were switched to a diet supplemented with C15:0 or linoleic acid (LA) for 11 weeks. A control group was fed with EFA during the whole study. Since linoleic acid deficiency is known to induce growth delay, weights were measured throughout the experiment and FA content in collected tissues were analyzed to evaluate biochemical markers of the deficiency. As expected, EFA-deficient rats showed growth retardation, compared to control rats. Supplementation of C15:0 at weaning increased early growth rate compared to deficient animals, as also did the supplementation of C18:2 n-6. Furthermore, the supplementation of C15:0 in the diet of EFA-deficient animals induced the previously undescribed synthesis of odd-chain PUFAs of the n-8 family (C19:3, C21:3 and C21:4 n-8). These results suggest dietary C15:0 might counteract EFA induced growth retardation, possibly through the synthesis of odd-chain n-8 PUFAs, yet mechanisms are to be deciphered for further validation.
Collapse
Affiliation(s)
- Vincent Ciesielski
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Thomas Guerbette
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Léa Fret
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France
| | - Mélodie Succar
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France
| | - Youenn Launay
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Patrice Dahirel
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Philippe Legrand
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Manuel Vlach
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Sophie Blat
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France
| | - Vincent Rioux
- Laboratory of Biochemistry and Human Nutrition, Institut Agro Rennes-Angers, Rennes, Bretagne, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, Bretagne, France.
| |
Collapse
|
11
|
Fomenko I, Denysenko N, Lozynska I, Kuryk M, Yushyn I, Myhal O, Pinyazhko R, Lozynskyi A, Lesyk R. Efficacy of thiazole derivatives against colorectal cancer induced by dimethylhydrazine in male Wistar rats. Biochem Biophys Res Commun 2025; 750:151424. [PMID: 39892058 DOI: 10.1016/j.bbrc.2025.151424] [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: 11/01/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Identifying novel biological activities in hit compounds remains a significant challenge in cancer research. In this study, we evaluated the anticancer and safety profiles of two previously studied thiazole-based derivatives, Les-5303 and Les-6485, in a colorectal cancer (CRC) model induced by dimethylhydrazine (DMH) in male Wistar rats. CRC was induced through subcutaneous DMH administration at a 20 mg/kg dose for 20 weeks. Les-5303 and Les-6485 were then administered intrarectally at a 6 mg/kg dose for 5 days. The effects of these compounds on oxidative stress, the antioxidant system, inflammation, NO-synthase activity in the colonic mucosa and blood plasma, and hepatotoxicity markers, were thoroughly assessed. Both compounds demonstrated significant anticancer activity and antioxidant properties. Les-5303, however, exhibited increased hepatotoxicity, as evidenced by elevated AST activity and reduced urea concentration in the blood, indicating its potential for liver damage. In contrast, Les-6485 showed no significant hepatotoxic effects, maintaining normal hepatic enzyme activities and urea levels, suggesting a better safety profile. These findings highlight the distinct biological effects of Les-5303 and Les-6485, with Les-6485 showing promising anticancer activity coupled with minimal toxicity, making it a more favorable candidate for further development in CRC treatment.
Collapse
Affiliation(s)
- Iryna Fomenko
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Nataliia Denysenko
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Iryna Lozynska
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Mariana Kuryk
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Ihor Yushyn
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Ostap Myhal
- Department of Therapeutic Stomatology, Periodontology and Stomatology FPGE, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Pinyazhko
- Department of Normal Physiology, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Andrii Lozynskyi
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine; Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszow, Poland.
| |
Collapse
|
12
|
Jiang B, Chen B, Xu J, Luo J, Yao M. Focused Electric Field Technology: A Novel Myoelectrical Stimulation Technology for Noninvasive Aging Muscle Rejuvenation. J Cosmet Dermatol 2025; 24:e16749. [PMID: 39719683 PMCID: PMC11845949 DOI: 10.1111/jocd.16749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/12/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Myoelectrical stimulation improves muscle function and reduces muscle atrophy and aging. However, research on the mechanism underlying its cosmetic effect remains limited. AIMS The aim of this study was to evaluate the cosmetic effects of the myoelectrical stimulation provided by the wearable intelligent flexible beauty device and its focused electric field technology (FEFT) on facial skin and muscle rejuvenation. PATIENTS/METHODS We conducted a single-blind, randomized, self-controlled clinical efficacy experiment on 31 female volunteers using the device. Using an FEFT-based platform, mice with d-galactose-induced skeletal muscle aging were subjected to surface myoelectrical stimulation of the gastrocnemius. Immunohistochemical analysis of skeletal muscles and protein immunoblotting were used to analyze the effects of FEFT. RESULTS After 14 days of use, facial skin elasticity significantly increased, wrinkle firmness significantly decreased, and the lift height of the upper eyelid and eye corner angle significantly increased in the volunteers. Clinical evaluation showed improvements in the drooping of the upper eyelid and eye bags. Self-evaluation questionnaires indicated alleviation of facial wrinkles. These improvements were more pronounced after 28 days. In mice, FEFT alleviated aging-induced muscle fiber atrophy, muscle fiber cross-sectional area reduction, and muscle satellite cell loss. FEFT also increased the expression of myogenic factors, including myogenic differentiation 1 (MYOD1). CONCLUSIONS FEFT exerted a skin-tightening effect by initiating myogenic processes and increasing the transformation of muscle satellite cells. Our research promotes the development of FEFT-based medical rehabilitation or cosmetic anti-aging products and provides a foundation for further application and comprehensive efficacy evaluation in human clinical settings.
Collapse
Affiliation(s)
- Boyang Jiang
- FLOSSOM Shenzhen Research InstituteShenzhenChina
- Shenzhen Rawskin DermatologyShenzhenChina
| | | | - Jia Xu
- FLOSSOM Shenzhen Research InstituteShenzhenChina
| | - Jieshu Luo
- Shenzhen Rawskin DermatologyShenzhenChina
| | - Min Yao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine ChinaShanghaiChina
| |
Collapse
|
13
|
Benak D, Sevcikova A, Holzerova K, Hlavackova M. FTO in health and disease. Front Cell Dev Biol 2024; 12:1500394. [PMID: 39744011 PMCID: PMC11688314 DOI: 10.3389/fcell.2024.1500394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Fat mass and obesity-associated (FTO) protein, a key enzyme integral to the dynamic regulation of epitranscriptomic modifications in RNAs, significantly influences crucial RNA lifecycle processes, including splicing, export, decay, and translation. The role of FTO in altering the epitranscriptome manifests across a spectrum of physiological and pathological conditions. This review aims to consolidate current understanding regarding the implications of FTO in health and disease, with a special emphasis on its involvement in obesity and non-communicable diseases associated with obesity, such as diabetes, cardiovascular disease, and cancer. It also summarizes the established molecules with FTO-inhibiting activity. Given the extensive impact of FTO on both physiology and pathophysiology, this overview provides illustrative insights into its roles, rather than an exhaustive account. A proper understanding of FTO function in human diseases could lead to new treatment approaches, potentially unlocking novel avenues for addressing both metabolic disorders and malignancies. The evolving insights into FTO's regulatory mechanisms hold great promise for future advancements in disease treatment and prevention.
Collapse
Affiliation(s)
| | | | | | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
14
|
Zaimoglu M, Secinti KD, Altinoz MA, Bozkurt M, Eroglu U, Ozpiskin O, Mammadkhanli O, Bayatli E, Caglar YS, Attar A. Organelle-level toxicity of nanometals relevant to titanium implants. Original research and comprehensive literature overview. Tissue Cell 2024; 91:102612. [PMID: 39546971 DOI: 10.1016/j.tice.2024.102612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study analyzed organelle toxicities of nanometals applied as free formulations or titanium rod-coating materials in rats. METHODS All materials were injected intraperitoneally, including the physiological saline applied to the control group. The first experimental group was implanted with nanosilver-coated titanium rods, and the second, third, and fourth groups received free nanosilver at rising levels. The fifth group was implanted with nanosilver, nanocopper, and nanozinc-coated titanium rods, and the sixth group received the same nanometals as free formulations. Light and electron microscopy and ICP-Mass Spectrometry were utilized to determine the neural, hepatic, and renal toxicities and tissue metal levels. RESULTS In brains, neuropil, myelin, and cellular damages occurred, especially in groups receiving high-dose nanosilver or nanometal combinations. Histiocyte accumulation and dark mitochondria within hepatocytes were discernible in the liver. Kidneys were the organs that were most severely affected by nanometal toxicity. The nephrotoxicity was apparent with the perturbations of the membrane infoldings and mitochondrial damage in the proximal and distal convoluted epithelia. Large angular peroxisomes developed inside the mesangial cells, and Golgi bodies increased in epithelial cells. Systemic metal levels increased on the thirtieth and prominently dropped on the sixtieth day. CONCLUSION These results provide insights into the extent of injury and organelle targets of nanometals and will guide optimizing the nanomaterials and implants used in the surgical practice.
Collapse
Affiliation(s)
- Murat Zaimoglu
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | - Kutsal Devrim Secinti
- Department of Neurosurgery, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Meric A Altinoz
- Department of Medical Biochemistry, Acibadem University, Altunizade, Istanbul, Turkey
| | - Melih Bozkurt
- Department of Neurosurgery, Istanbul Arel University, Istanbul, Turkey; Department of Neurosurgery, Memorial Bahcelievler Hospital, Memorial Health Group, Istanbul, Turkey
| | - Umit Eroglu
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Omer Ozpiskin
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Orkhan Mammadkhanli
- Department of Neurosurgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Eyup Bayatli
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Yusuf Sukru Caglar
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ayhan Attar
- Department of Neurosurgery, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
15
|
Kearns ML, Reynolds CM. The impact of non-nutritive sweeteners on fertility, maternal and child health outcomes: a review of human and animal studies. Proc Nutr Soc 2024; 83:280-292. [PMID: 38433591 DOI: 10.1017/s0029665124000168] [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: 03/05/2024]
Abstract
There is significant evidence that an unhealthy diet greatly increases the risk of complications during pregnancy and predisposes offspring to metabolic dysfunction and obesity. While fat intake is typically associated with the onset of obesity and its comorbidities, there is increasing evidence linking sugar, particularly high fructose corn syrup, to the global rise in obesity rates. Furthermore, the detrimental effects of added sugar intake during pregnancy on mother and child have been clearly outlined. Guidelines advising pregnant women to avoid food and beverages with high fat and sugar have led to an increase in consumption of 'diet' or 'light' options. Examination of some human birth cohort studies shows that heavy consumption (at least one beverage a day) of non-nutritive sweetener (NNS) containing beverages has been associated with increased risk of preterm birth and increased weight/BMI in male offspring independent of maternal weight, which appears to be offset by breastfeeding for 6 months. Rodent models have shown that NNS exposure during pregnancy can impact maternal metabolic health, adipose tissue function, gut microbiome profiles and taste preference. However, the mechanisms underlying these effects are multifaceted and further research, particularly in a translational setting is required to fully understand the effects of NNS on maternal and infant health during pregnancy. Therefore, this review examines maternal sweetener intakes and their influence on fertility, maternal health outcomes and offspring outcomes in human cohort studies and rodent models.
Collapse
Affiliation(s)
- Michelle L Kearns
- School of Public Health, Physiotherapy and Sports Science/Conway Institute/Institute of Food and Health/Diabetes Complications Research Centre, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - Clare M Reynolds
- School of Public Health, Physiotherapy and Sports Science/Conway Institute/Institute of Food and Health/Diabetes Complications Research Centre, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
16
|
May HG, Tsikonofilos K, Donat CK, Sastre M, Kozlov AS, Sharp DJ, Bruyns-Haylett M. EEG hyperexcitability and hyperconnectivity linked to GABAergic inhibitory interneuron loss following traumatic brain injury. Brain Commun 2024; 6:fcae385. [PMID: 39605970 PMCID: PMC11600960 DOI: 10.1093/braincomms/fcae385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Traumatic brain injury represents a significant global health burden and has the highest prevalence among neurological disorders. Even mild traumatic brain injury can induce subtle, long-lasting changes that increase the risk of future neurodegeneration. Importantly, this can be challenging to detect through conventional neurological assessment. This underscores the need for more sensitive diagnostic tools, such as electroencephalography, to uncover opportunities for therapeutic intervention. Progress in the field has been hindered by a lack of studies linking mechanistic insights at the microscopic level from animal models to the macroscale phenotypes observed in clinical imaging. Our study addresses this gap by investigating a rat model of mild blast traumatic brain injury using both immunohistochemical staining of inhibitory interneurons and translationally relevant electroencephalography recordings. Although we observed no pronounced effects immediately post-injury, chronic time points revealed broadband hyperexcitability and increased connectivity, accompanied by decreased density of inhibitory interneurons. This pattern suggests a disruption in the balance between excitation and inhibition, providing a crucial link between cellular mechanisms and clinical hallmarks of injury. Our findings have significant implications for the diagnosis, monitoring, and treatment of traumatic brain injury. The emergence of electroencephalography abnormalities at chronic time points, despite the absence of immediate effects, highlights the importance of long-term monitoring in traumatic brain injury patients. The observed decrease in inhibitory interneuron density offers a potential cellular mechanism underlying the electroencephalography changes and may represent a target for therapeutic intervention. This study demonstrates the value of combining cellular-level analysis with macroscale neurophysiological recordings in animal models to elucidate the pathophysiology of traumatic brain injury. Future research should focus on translating these findings to human studies and exploring potential therapeutic strategies targeting the excitation-inhibition imbalance in traumatic brain injury.
Collapse
Affiliation(s)
- Hazel G May
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Konstantinos Tsikonofilos
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
- Department of Medicinal Radiochemistry, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Andriy S Kozlov
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - David J Sharp
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Michael Bruyns-Haylett
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Bioengineering, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
- Department of Quantitative Methods, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
| |
Collapse
|
17
|
Jia Y, Wei Z, Feng J, Lei M, Yang Y, Liu J, Ma Y, Chen W, Huang G, Genin GM, Guo X, Li Y, Xu F. A Heart Rate Matched Patch for Mechano-Chemical Treatment of Myocardial Infarction: Optimal Design and Transspecies Application. RESEARCH (WASHINGTON, D.C.) 2024; 7:0517. [PMID: 39582687 PMCID: PMC11582187 DOI: 10.34133/research.0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 11/26/2024]
Abstract
After myocardial infarction (MI), ventricular dilation and the microscopic passive stretching of the infarcted border zone is the meaning contributor to the continuous expansion of myocardial fibrosis. Epicardial hydrogel patches have been demonstrated to alleviate this sequela of MI in small-animal models. However, these have not been successfully translated to humans or even large animals, in part because of challenges in attaining both the greater stiffness and slower viscoelastic relaxation that mathematical models predict to be optimal for application to larger, slower-beating hearts. Here, using borate-based dynamic covalent chemistry, we develop an injectable "heart rate matched" viscoelastic gelatin (VGtn) hydrogel with a gel point tunable across the stiffnesses and frequencies that are predicted to transspecies and cross-scale cardiac repair after MI. Small-animal experiments demonstrated that, compared to heart rate mismatched patches, the heart rate matched VGtn patches inhibited ventricular bulging and attenuated stress concentrations in the myocardium after MI. In particular, the viscoelastic patch can coordinate the microscopic strain at the infarction boundary. VGtn loaded with anti-fibrotic agents further reduced myocardial damage and promoted angiogenesis in the myocardium. The tuned heart rate matched patches demonstrated similar benefits in a larger-scale and lower heart rate porcine MI model. Results suggest that heart rate matched VGtn patches may hold potential for clinical translation.
Collapse
Affiliation(s)
- Yuanbo Jia
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, P.R. China
- Key Laboratory of Surgical Critical Care and Life Support (Xi’an Jiaotong University), Ministry of Education, Xi’an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Zhao Wei
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Jinteng Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Meng Lei
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Yanshen Yang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Jingyi Liu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Yufei Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Weiguo Chen
- Department of Cardiology, Tangdu Hospital,
the Air Force Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering,
Wuhan University, Wuhan 430072, P.R. China
| | - Guy M. Genin
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Department of Mechanical Engineering & Materials Science,
Washington University in St. Louis, St. Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology,
Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou 310003, P.R. China
| | - Yan Li
- Department of Cardiology, Tangdu Hospital,
the Air Force Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| |
Collapse
|
18
|
Kamar SA, Naiem Hamdy K, El-Nefiawy NE, Mohammed H, Fetouh MA. Exploring Mesenchymal Stem Cells versus Minoxidil for Androgenic Alopecia Treatment: A Detailed Animal-Based Histological and Morphometric Study. Cells Tissues Organs 2024:1-17. [PMID: 39527940 DOI: 10.1159/000542547] [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: 07/02/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Androgenic alopecia (AGA), a hair loss condition caused by dihydrotestosterone binding to hair follicle receptors, negatively impacts quality of life for both men and women. Current treatments like minoxidil and finasteride have limitations, highlighting the need for alternative therapies, such as human umbilical cord blood-derived mesenchymal stem cells (HUCB-MSCs). METHODS In this study, forty-eight adult male Wistar albino rats (3 months old) were used. The control group (Group I) received no treatment, while the other rats underwent AGA induction via daily subcutaneous testosterone injections (100 mg/kg). These rats developed alopecia and were divided into three groups: AGA (Group II), AGA plus daily minoxidil spray (Group III), and AGA plus a single intradermal injection of HUCB-MSCs (1 mL containing 1 × 105 cells, Group IV). After 4 weeks, the rats were sacrificed, and skin specimens were prepared for histological analysis using H&E, Masson's trichrome, and immunohistochemical staining for CK 19, vascular endothelial growth factor (VEGF), and TUNEL antibodies. RESULTS It was shown that HUCB-MSC treatment reversed structural damage to hair and follicles, normalizing conditions within 1-week post-injection. The treatment enhanced the anagen phase, suppressed telogen and catagen phases, reduced apoptosis, and increased VEGF and CK 19 immune reactions. Observational follow-up for Groups III and IV revealed that while the minoxidil group experienced significant hair loss after 37 days, the stem cell group exhibited dense and long hair covering the treated area. CONCLUSION HUCB-MSC therapy demonstrated superior efficacy over minoxidil with no observed side effects, indicating its potential as a promising alternative for AGA treatment.
Collapse
Affiliation(s)
- Sherif A Kamar
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
| | - Khaled Naiem Hamdy
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Heba Mohammed
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa A Fetouh
- Department Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
19
|
Caruana V, Giles BH, Kukolj N, Juran R, Baglole CJ, Mann KK. Chronic exposure to E-cigarette aerosols potentiates atherosclerosis in a sex-dependent manner. Toxicol Appl Pharmacol 2024; 492:117095. [PMID: 39245079 DOI: 10.1016/j.taap.2024.117095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Despite being designed for smoking cessation, e-cigarettes and their variety of flavors have become increasingly attractive to teens and young adults. This trend has fueled concerns regarding the potential role of e-cigarettes in advancing chronic diseases, notably those affecting the cardiovascular system. E-cigarettes contain a mixture of metals and chemical compounds, some of which have been implicated in cardiovascular diseases like atherosclerosis. Our laboratory has optimized in vivo exposure regimens to mimic human vaping patterns. Using these established protocols in an inducible (AAV-PCSK9) hyperlipidemic mouse model, this study tests the hypothesis that a chronic exposure to e-cigarette aerosols will increase atherosclerotic plaques. The exposures were conducted using the SCIREQ InExpose™ nose-only inhalation system and STLTH or Vuse products for 16 weeks. We observed that only male mice exposed to STLTH or Vuse aerosols had significantly increased plasma total cholesterol, triglycerides, and LDL cholesterol levels compared to mice exposed to system air. Moreover, these male mice also had a significant increase in aortic and sinus plaque area. Male mice exposed to e-cigarette aerosol had a significant reduction in weight gain over the exposure period. Our data indicate that e-cigarette use in young hyperlipidemic male mice increases atherosclerosis in the absence of significant pulmonary and systemic inflammation. These results underscore the need for extensive research to unravel the long-term health effects of e-cigarettes.
Collapse
Affiliation(s)
- Vincenza Caruana
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Braeden H Giles
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Nikola Kukolj
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Roni Juran
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Carolyn J Baglole
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Koren K Mann
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada.
| |
Collapse
|
20
|
Gawrys O, Jíchová Š, Miklovič M, Husková Z, Kikerlová S, Sadowski J, Kollárová P, Lenčová-Popelova O, Hošková L, Imig JD, Mazurova Y, Kolář F, Melenovský V, Štěrba M, Červenka L. Characterization of a new model of chemotherapy-induced heart failure with reduced ejection fraction and nephrotic syndrome in Ren-2 transgenic rats. Hypertens Res 2024; 47:3126-3146. [PMID: 39245782 PMCID: PMC11534684 DOI: 10.1038/s41440-024-01865-7] [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: 04/16/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024]
Abstract
All anthracyclines, including doxorubicin (DOXO), the most common and still indispensable drug, exhibit cardiotoxicity with inherent risk of irreversible cardiomyopathy leading to heart failure with reduced ejection fraction (HFrEF). Current pharmacological strategies are clearly less effective for this type of HFrEF, hence an urgent need for new therapeutic approaches. The prerequisite for success is thorough understanding of pathophysiology of this HFrEF form, which requires an appropriate animal model of the disease. The aim of this study was to comprehensively characterise a novel model of HF with cardiorenal syndrome, i.e. DOXO-induced HFrEF with nephrotic syndrome, in which DOXO was administered to Ren-2 transgenic rats (TGR) via five intravenous injections in a cumulative dose of 10 mg/kg of body weight (BW). Our analysis included survival, echocardiography, as well as histological examination of the heart and kidneys, blood pressure, but also a broad spectrum of biomarkers to evaluate cardiac remodelling, fibrosis, apoptosis, oxidative stress and more. We have shown that the new model adequately mimics the cardiac remodelling described as "eccentric chamber atrophy" and myocardial damage typical for DOXO-related cardiotoxicity, without major damage of the peritoneum, lungs and liver. This pattern corresponds well to a clinical situation of cancer patients receiving anthracyclines, where HF develops with some delay after the anticancer therapy. Therefore, this study may serve as a comprehensive reference for all types of research on DOXO-related cardiotoxicity, proving especially useful in the search for new therapeutic strategies.
Collapse
Affiliation(s)
- Olga Gawrys
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Šárka Jíchová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Matúš Miklovič
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Zuzana Husková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Janusz Sadowski
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petra Kollárová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Olga Lenčová-Popelova
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Lenka Hošková
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - John D Imig
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yvona Mazurova
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - František Kolář
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic.
| |
Collapse
|
21
|
da Veiga Dutra ML, de Souza DM, Santos HMCC, Cruz Neto JPR, Soares NL, Vieira ACA, Costa IKC, Rolim TBB, de Magalhães Cordeiro ÂMT, de Vasconcelos DAA, Lira EC, Alves AF, Aquino JDS. Effects of maternal preconception high-fat diet on the fertility of dams and on the somatic parameters and reflex ontogeny of their male offspring. Physiol Behav 2024; 288:114723. [PMID: 39481508 DOI: 10.1016/j.physbeh.2024.114723] [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: 06/07/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND Female consumption of a high-fat diet (HFD) may cause fertility issues and affects offspring development. OBJECTIVE Evaluate the acute maternal preconception intake of a HFD on the fertility and reproduction parameters of breeding females; and on the somatic parameters and reflex ontogeny of male offspring. METHODS Twenty-four rats were randomized into control (PC; n=12) and high-fat diet group (PHF; n=12) that consumed their respective diets during the 23-day preconception period. After that, 6 rats per group underwent oral glucose tolerance and insulin tolerance tests and were euthanized. The remaining rats were mated, during gestation and lactation, both groups ate a control diet. After birth, the male offspring's somatic parameters and reflexes were assessed. RESULTS The preconception diet caused dyslipidemia in the PHF. The PHF uterus exhibited a higher SFA (50.74 ± 0.32 %), a lower PUFA concentration (35.59 ± 0.33 %), and an increase in arachidonic acid (2.48 ± 0.03 %). PHF rats had hypertrophy in the endometrium, and ovaries with a higher quantity of corpora albicans and immature primordial follicles. The offspring of PHF rats had greater weight (6.70 ± 0.82 g), nasal-anal length (4.93± 0.27 cm), and tail length (1.74 ± 0.12 cm) on the first day of life, and had improved righting reflex, but delayed negative geotaxis reflex. CONCLUSIONS An acute maternal preconception HFD induced a pro-inflammatory fatty acid profile and changed structure in uterus, altered ovarian follicle profile. Also, potential interference in the size of the pups at birth and in brain development of male offspring.
Collapse
Affiliation(s)
- Maria Letícia da Veiga Dutra
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Danielle Melo de Souza
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Harley Mateus Coutinho Correia Santos
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - José Patrocínio Ribeiro Cruz Neto
- Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Naís Lira Soares
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Anne Caroline Alves Vieira
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Isabelle Karoline Carvalho Costa
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Thaís Bayma Barbosa Rolim
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | | | - Diogo Antônio Alves de Vasconcelos
- Phisiology Laboratory, Department of Nutrition, Health Sciences Centre, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Eduardo Carvalho Lira
- Neuroendocrinology and Metabolism Laboratory, Department of Physiology and Pharmacology, Health Sciences Centre, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Adriano Francisco Alves
- Laboratory of General pathology, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Jailane de Souza Aquino
- Laboratory of Experimental Nutrition, Department of Nutrition, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Nutrition Sciences, Health Sciences Centre, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil.
| |
Collapse
|
22
|
Giuliani C, Di Dalmazi G, Bucci I, Napolitano G. Quercetin and Thyroid. Antioxidants (Basel) 2024; 13:1202. [PMID: 39456456 PMCID: PMC11505551 DOI: 10.3390/antiox13101202] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Quercetin is the most abundant flavonoid present in fruits and vegetables. For its antiproliferative, antiviral, anti-inflammatory and antioxidants activities, it is an active ingredient of several herbal remedies and is available as a nutraceutical. Experimental studies performed in vitro have demonstrated that quercetin inhibits growth and function in normal thyroid cells and may act as a thyroid disruptor. These effects have also been confirmed in vivo using rodent models. Some studies have reported the ability of quercetin to interfere with the metabolism of thyroid hormones, since it inhibits the 5'-deiodinase type 1 (D1) activity in the thyroid, as well as in the liver. Besides the effects on normal thyroid cells, several experiments performed in vitro have shown a potential therapeutic role of quercetin in thyroid cancer. Indeed, quercetin inhibits the growth, the adhesion and the migration of thyroid cancer cells, and it also has redifferentiation properties in some thyroid cancer cell lines. In conclusion, these data suggest that, although its effects can be of benefit in hyperthyroidism and thyroid cancer, caution is required in the use of high doses of quercetin due to its anti-thyroid properties. Further in vivo studies are certainly needed to confirm these hypotheses.
Collapse
Affiliation(s)
- Cesidio Giuliani
- Unit of Endocrinology, Department of Medicine and Sciences of Aging and Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy; (G.D.D.); (I.B.); (G.N.)
| | | | | | | |
Collapse
|
23
|
Jang S, Lee JB, Yoo C, Kim HS, Choi K, Lee J, Lee DY. Biocompatible and nondegradable microcapsules using an ethylamine-bridged EGCG dimer for successful therapeutic cell transplantation. J Control Release 2024; 373:520-532. [PMID: 39059498 DOI: 10.1016/j.jconrel.2024.07.053] [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: 02/23/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Conventional alginate microcapsules are widely used for encapsulating therapeutic cells to reduce the host immune response. However, the exchange of monovalent cations with divalent cations for crosslinking can lead to a sol-gel phase transition, resulting in gradual degradation and swelling of the microcapsules in the body. To address this limitation, we present a biocompatible and nondegradable epigallocatechin-3-gallate (EGCG)-based microencapsulation with ethylamine-bridged EGCG dimers (EGCG(d)), denoted as 'Epi-Capsules'. These Epi-Capsules showed increased physical properties and Ca2+ chelating resistance compared to conventional alginate microcapsules. Horseradish peroxidase (HRP) treatment is very effective in increasing the stability of Epi-Capsule((+)HRP) due to the crosslinking between EGCG(d) molecules. Interestingly, the Epi-Capsules(oxi) using a pre-oxidized EGCG(d) can support long-term survival (>90 days) of xenotransplanted insulin-secreting islets in diabetic mice in vivo, which is attributed to its structural stability and reactive oxygen species (ROS) scavenging for lower fibrotic activity. Collectively, this EGCG-based microencapsulation can create Ca2+ chelating-resistance and anti-oxidant activity, which could be a promising strategy for cell therapies for diabetes and other diseases.
Collapse
Affiliation(s)
- Seonmi Jang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Jae Bin Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Chaerim Yoo
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyung Shik Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Kimyung Choi
- Optipharm Co., Ltd., Cheongju 28158, Republic of Korea
| | - Joonseok Lee
- Department of Chemistry, Hanyang University, Seoul 04763, Republic of Korea.
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul 04763, Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul 04763, Republic of Korea; Elixir Pharmatech Inc., Seoul 04763, Republic of Korea.
| |
Collapse
|
24
|
Fernández-Serra R, Lekouaghet A, Peracho L, Yonesi M, Alcázar A, Chioua M, Marco-Contelles J, Pérez-Rigueiro J, Rojo FJ, Panetsos F, Guinea GV, González-Nieto D. Permselectivity of Silk Fibroin Hydrogels for Advanced Drug Delivery Neurotherapies. Biomacromolecules 2024; 25:5233-5250. [PMID: 39018332 PMCID: PMC11323009 DOI: 10.1021/acs.biomac.4c00629] [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: 05/09/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 07/19/2024]
Abstract
A promising trend in tissue engineering is using biomaterials to improve the control of drug concentration in targeted tissue. These vehicular systems are of specific interest when the required treatment time window is higher than the stability of therapeutic molecules in the body. Herein, the capacity of silk fibroin hydrogels to release different molecules and drugs in a sustained manner was evaluated. We found that a biomaterial format, obtained by an entirely aqueous-based process, could release molecules of variable molecular weight and charge with a preferential delivery of negatively charged molecules. Although the theoretical modeling suggested that drug delivery was more likely to be driven by Fickian diffusion, the external media had a considerable influence on the release, with lipophilic organic solvents such as acetonitrile-methanol (ACN-MeOH) intensifying the release of hydrophobic molecules. Second, we found that silk fibroin could be used as a vehicular system to treat a variety of brain disorders as this biomaterial sustained the release of different factors with neurotrophic (brain-derived neurotrophic factor) (BDNF), chemoattractant (C-X-C motif chemokine 12) (CXCL12), anti-inflammatory (TGF-β-1), and angiogenic (VEGF) capacities. Finally, we demonstrated that this biomaterial hydrogel could release cholesteronitrone ISQ201, a nitrone with antioxidant capacity, showing neuroprotective activity in an in vitro model of ischemia-reoxygenation. Given the slow degradation rate shown by silk fibroin in many biological tissues, including the nervous system, our study expands the restricted list of drug delivery-based biomaterial systems with therapeutic capacity for both short- and especially long-term treatment windows and has merit for use with brain pathologies.
Collapse
Affiliation(s)
- Rocío Fernández-Serra
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
| | - Amira Lekouaghet
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
| | - Lorena Peracho
- Department
of Research, Hospital Universitario Ramón
y Cajal, Madrid 28034, Spain
- Proteomics
Unit, Instituto Ramón y Cajal de
Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Mahdi Yonesi
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
| | - Alberto Alcázar
- Department
of Research, Hospital Universitario Ramón
y Cajal, Madrid 28034, Spain
- Proteomics
Unit, Instituto Ramón y Cajal de
Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Mourad Chioua
- Laboratory
of Medicinal Chemistry, Institute of General
Organic Chemistry (CSIC), Madrid 28006, Spain
| | - José Marco-Contelles
- Laboratory
of Medicinal Chemistry, Institute of General
Organic Chemistry (CSIC), Madrid 28006, Spain
- Center
for
Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid 28029, Spain
| | - José Pérez-Rigueiro
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Francisco J. Rojo
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Fivos Panetsos
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
- Neurocomputing
and Neurorobotics Research Group, Faculty of Biology and Faculty of
Optics, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Gustavo V. Guinea
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Daniel González-Nieto
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Departamento de
Tecnología Fotónica y Bioingeniería,
ETSI Telecomunicaciones, Universidad Politécnica
de Madrid, Madrid 28040, Spain
| |
Collapse
|
25
|
Edinger F, Zajonz T, Mayer N, Schmidt G, Schneck E, Sander M, Koch C. A Novel Model of Venovenous Extracorporeal Membrane Oxygenation in Rats with Femoral Cannulation and Insights into Hemodynamic Changes. Biomedicines 2024; 12:1819. [PMID: 39200283 PMCID: PMC11351971 DOI: 10.3390/biomedicines12081819] [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: 07/16/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
The application of venovenous (VV) extracorporeal membrane oxygenation (ECMO) has gained wide acceptance for the treatment of acute severe respiratory failure. Since no rat model of VV ECMO therapy with femoral drainage has yet been described, although this cannulation strategy is commonly used in humans, this study aimed to establish such a model. Twenty male Lewis rats were randomly assigned to receive a sham procedure or VV ECMO therapy. After the inhalative induction of anesthesia, animals were intubated and the vascular accesses were placed surgically. While venous drainage was achieved through a modified multi-orifice 18 G cannula that was placed in the inferior vena cava through the femoral vein over a guide wire with an ultra-flexible tip, the venous return was realized via a shortened 20 G cannula into the jugular vein. Hemodynamic data were obtained from a tail artery and left ventricular pressure-volume catheter. Repetitive blood gas analyses were carried out, and systemic inflammation was measured using an enzyme-linked immunosorbent assay. While animals in the ECMO group showed adequate oxygenation and decarboxylation, there was no evidence of recirculation. VV ECMO therapy increased stroke volume (SV), cardiac output (CO), and left ventricular end-diastolic volume (LVEDV). ECMO-induced inflammation was reflected in increased levels of tumor necrosis factor alpha. However, no differences in interleukins 6 and 10 were seen. This study describes a frequently used cannulation strategy in humans for a rat model of VV ECMO. Despite successful oxygenation and decarboxylation, the oxygenated blood may reduce pulmonary vascular resistance and lead to an increased LVEDV, which is associated with increased SV and CO. This model allows us to answer research questions about topics such as intestinal microcirculation in further studies.
Collapse
Affiliation(s)
| | - Thomas Zajonz
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital, Justus-Liebig-University, 35392 Giessen, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Li D, Donnelley M, Parsons D, Habgood MD, Schneider-Futschik EK. Extent of foetal exposure to maternal elexacaftor/tezacaftor/ivacaftor during pregnancy. Br J Pharmacol 2024; 181:2413-2428. [PMID: 38770951 DOI: 10.1111/bph.16417] [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: 10/24/2023] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Cystic fibrosis (CF) patients are living longer and healthier due to improved treatments, e.g. cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy elexacaftor/tezacaftor/ivacaftor (ETI), with treatment possibly occurring in pregnancy. The risk of ETI to foetuses remain unknown. Thus the effect of maternally administered ETI on foetal genetic and structural development was investigated. EXPERIMENTAL APPROACH Pregnant Sprague Dawley rats were orally treated with ETI (6.7 mg·kg-1·day-1 elexacaftor + 3.5 mg·kg-1·day-1 tezacaftor + 25 mg·kg-1·day-1 ivacaftor) for 7 days from E12 to E19. Tissue samples collected at E19 were analysed using histology and RNA sequencing. Histological changes and differentially expressed genes (DEG) were assessed. KEY RESULTS No overt structural abnormalities were found in foetal pancreas, liver, lung and small intestine after 7-day ETI exposure. Very few non-functionally associated DEG in foetal liver, lung and small intestine were identified using RNA-seq. 29 DEG were identified in thymus (27 up-regulated and two down-regulated) and most were functionally linked to each other. Gene ontology enrichment analysis revealed that multiple muscle-related terms were significantly enriched. Many more DEG were identified in cortex (44 up-regulated and four down-regulated) and a group of these were involved in central nervous system and brain development. CONCLUSION AND IMPLICATION Sub-chronic ETI treatment in late pregnancy does not appear to pose a significant risk to the genetic and structural development of many foetal tissues. However, significant gene changes in foetal thymic myoid cells and cortical neuronal development requires future follow-up studies to assess the risk to these organs.
Collapse
Affiliation(s)
- Danni Li
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Mark D Habgood
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Elena K Schneider-Futschik
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
27
|
Xu J, Bao G, Jia B, Wang M, Wen P, Kan T, Zhang S, Liu A, Tang H, Yang H, Yue B, Dai K, Zheng Y, Qu X. An adaptive biodegradable zinc alloy with bidirectional regulation of bone homeostasis for treating fractures and aged bone defects. Bioact Mater 2024; 38:207-224. [PMID: 38756201 PMCID: PMC11096722 DOI: 10.1016/j.bioactmat.2024.04.027] [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: 03/01/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
Healing of fractures or bone defects is significantly hindered by overactivated osteoclasts and inhibited osteogenesis in patients with abnormal bone metabolism. Current clinical approaches using titanium alloys or stainless steel provide mechanical support but have no biological effects on bone regeneration. Therefore, designing and fabricating degradable metal materials with sufficient mechanical strength and bidirectional regulation of both osteoblasts and osteoclasts is a substantial challenge. Here, this study first reported an adaptive biodegradable Zn-0.8 Mg alloy with bidirectional regulation of bone homeostasis, which promotes osteogenic differentiation by activating the Pi3k/Akt pathway and inhibits osteoclast differentiation by inhibiting the GRB2/ERK pathway. The anti-osteolytic ability of the Zn-0.8 Mg alloy was verified in a mouse calvarial osteolysis model and its suitability for internal fracture fixation with high-strength screws was confirmed in the rabbit femoral condyle fracture model. Furthermore, in an aged postmenopausal rat femoral condyle defect model, 3D printed Zn-0.8 Mg scaffolds promoted excellent bone regeneration through adaptive structures with good mechanical properties and bidirectionally regulated bone metabolism, enabling personalized bone defect repair. These findings demonstrate the substantial potential of the Zn-0.8 Mg alloy for treating fractures or bone defects in patients with aberrant bone metabolism.
Collapse
Affiliation(s)
- Jialian Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Guo Bao
- Laboratory Animal centre, National Research Institute for Family Planning, Beijing, 100081, China
| | - Bo Jia
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Tianyou Kan
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Aobo Liu
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Kerong Dai
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| |
Collapse
|
28
|
Li C, Qian W, Wei X, Narasimhan H, Wu Y, Arish M, Cheon IS, Tang J, de Almeida Santos G, Li Y, Sharifi K, Kern R, Vassallo R, Sun J. Comparative single-cell analysis reveals IFN-γ as a driver of respiratory sequelae after acute COVID-19. Sci Transl Med 2024; 16:eadn0136. [PMID: 39018367 DOI: 10.1126/scitranslmed.adn0136] [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: 11/17/2023] [Revised: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/19/2024]
Abstract
Postacute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (PASC) represent an urgent public health challenge and are estimated to affect more than 60 million individuals globally. Although a growing body of evidence suggests that dysregulated immune reactions may be linked with PASC symptoms, most investigations have primarily centered around blood-based studies, with few focusing on samples derived from affected tissues. Furthermore, clinical studies alone often provide correlative insights rather than causal mechanisms. Thus, it is essential to compare clinical samples with relevant animal models and conduct functional experiments to understand the etiology of PASC. In this study, we comprehensively compared bronchoalveolar lavage fluid single-cell RNA sequencing data derived from clinical PASC samples and a mouse model of PASC. This revealed a pro-fibrotic monocyte-derived macrophage response in respiratory PASC, as well as abnormal interactions between pulmonary macrophages and respiratory resident T cells, in both humans and mice. Interferon-γ (IFN-γ) emerged as a key node mediating the immune anomalies in respiratory PASC. Neutralizing IFN-γ after the resolution of acute SARS-CoV-2 infection reduced lung inflammation and tissue fibrosis in mice. Together, our study underscores the importance of performing comparative analysis to understand the cause of PASC and suggests that the IFN-γ signaling axis might represent a therapeutic target.
Collapse
Affiliation(s)
- Chaofan Li
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Xiaoqin Wei
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Harish Narasimhan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yue Wu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Mohd Arish
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - In Su Cheon
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jinyi Tang
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Gislane de Almeida Santos
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ying Li
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kamyar Sharifi
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ryan Kern
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert Vassallo
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
29
|
Carles L, Gibaja A, Scheper V, Alvarado JC, Almodovar C, Lenarz T, Juiz JM. Efficacy and Mechanisms of Antioxidant Compounds and Combinations Thereof against Cisplatin-Induced Hearing Loss in a Rat Model. Antioxidants (Basel) 2024; 13:761. [PMID: 39061830 PMCID: PMC11273477 DOI: 10.3390/antiox13070761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Cisplatin is an election chemotherapeutic agent used for many cancer treatments. Its cytotoxicity against neoplastic cells is mirrored by that taking place in healthy cells and tissues, resulting in serious adverse events. A very frequent one is ototoxicity, causing hearing loss which may permanently affect quality of life after successful oncologic treatments. Exacerbated oxidative stress is a main cytotoxic mechanism of cisplatin, including ototoxicity. Previous reports have shown antioxidant protection against cisplatin ototoxicity, but there is a lack of comparative studies on the otoprotectant activity and mechanism of antioxidant formulations. Here, we show evidence that a cocktail of vitamins A, C, and E along with Mg++ (ACEMg), previously shown to protect against noise-induced hearing loss, reverses auditory threshold shifts, promotes outer hair cell survival, and attenuates oxidative stress in the cochlea after cisplatin treatment, thus protecting against extreme cisplatin ototoxicity in rats. The addition of 500 mg N-acetylcysteine (NAC), which, administered individually, also shows significant attenuation of cisplatin ototoxicity, to the ACEMg formulation results in functional degradation of ACEMg otoprotection. Mg++ administered alone, as MgSO4, also prevents cisplatin ototoxicity, but in combination with 500 mg NAC, otoprotection is also greatly degraded. Increasing the dose of NAC to 1000 mg also results in dramatic loss of otoprotection activity compared with 500 mg NAC. These findings support that single antioxidants or antioxidant combinations, particularly ACEMg in this experimental series, have significant otoprotection efficacy against cisplatin ototoxicity. However, an excess of combined antioxidants and/or elevated doses, above a yet-to-be-defined "antioxidation threshold", results in unrecoverable redox imbalance with loss of otoprotectant activity.
Collapse
Grants
- PID2020-117266RB-C22-1, EXC 2177/1, ID:390895286, SBPLY/17/180501/000544. Ministerio de Ciencia Innovación, MCINN, Gobierno de España, Plan Estatal de I+D+i, PID2020-117266RB-C22-1, Cluster of Excellence "Hearing4All" EXC 2177/1, ID:390895286, part of the Germany´s Excellence Strategy of the German Research Foundation, DFG. Co
Collapse
Affiliation(s)
- Liliana Carles
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha (UCLM), Campus in Albacete, 02008 Albacete, Spain; (L.C.); (A.G.); (J.C.A.)
- Department of Otolaryngology, University Hospital “Doce de Octubre”, 28041 Madrid, Spain;
| | - Alejandro Gibaja
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha (UCLM), Campus in Albacete, 02008 Albacete, Spain; (L.C.); (A.G.); (J.C.A.)
| | - Verena Scheper
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (V.S.); (T.L.)
- Cluster of Excellence “Hearing4all” of the German Research Foundation, DFG, 26111 Oldenburg, Germany
| | - Juan C. Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha (UCLM), Campus in Albacete, 02008 Albacete, Spain; (L.C.); (A.G.); (J.C.A.)
| | - Carlos Almodovar
- Department of Otolaryngology, University Hospital “Doce de Octubre”, 28041 Madrid, Spain;
| | - Thomas Lenarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (V.S.); (T.L.)
- Cluster of Excellence “Hearing4all” of the German Research Foundation, DFG, 26111 Oldenburg, Germany
| | - José M. Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha (UCLM), Campus in Albacete, 02008 Albacete, Spain; (L.C.); (A.G.); (J.C.A.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (V.S.); (T.L.)
- Cluster of Excellence “Hearing4all” of the German Research Foundation, DFG, 26111 Oldenburg, Germany
| |
Collapse
|
30
|
Aliena-Valero A, Hernández-Jiménez M, López-Morales MA, Tamayo-Torres E, Castelló-Ruiz M, Piñeiro D, Ribó M, Salom JB. Cerebroprotective Effects of the TLR4-Binding DNA Aptamer ApTOLL in a Rat Model of Ischemic Stroke and Thrombectomy Recanalization. Pharmaceutics 2024; 16:741. [PMID: 38931862 PMCID: PMC11206667 DOI: 10.3390/pharmaceutics16060741] [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: 04/24/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
ApTOLL, a TLR4 modulator aptamer, has demonstrated cerebroprotective effects in a permanent ischemic stroke mouse model, as well as safety and efficacy in early phase clinical trials. We carried out reverse translation research according to STAIR recommendations to further characterize the effects and mechanisms of ApTOLL after transient ischemic stroke in rats and to better inform the design of pivotal clinical trials. Adult male rats subjected to transient middle cerebral artery occlusion were treated either with ApTOLL or the vehicle intravenously at different doses and time-points. ApTOLL was compared with TAK-242 (a TLR4 inhibitor). Female rats were also studied. After neurofunctional evaluation, brains were removed for infarct/edema volume, hemorrhagic transformation, and histologic determinations. Peripheral leukocyte populations were assessed via flow cytometry. ApTOLL showed U-shaped dose-dependent cerebroprotective effects. The maximum effective dose (0.45 mg/kg) was cerebroprotective when given both before reperfusion and up to 12 h after reperfusion and reduced the hemorrhagic risk. Similar effects occurred in female rats. Both research and clinical ApTOLL batches induced slightly superior cerebroprotection when compared with TAK-242. Finally, ApTOLL modulated circulating leukocyte levels, reached the brain ischemic tissue to bind resident and infiltrated cell types, and reduced the neutrophil density. These results show the cerebroprotective effects of ApTOLL in ischemic stroke by reducing the infarct/edema volume, neurofunctional impairment, and hemorrhagic risk, as well as the peripheral and local immune response. They provide information about ApTOLL dose effects and its therapeutic time window and target population, as well as its mode of action, which should be considered in the design of pivotal clinical trials.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.A.-V.); (M.A.L.-M.); (M.C.-R.)
| | - Macarena Hernández-Jiménez
- AptaTargets S.L., 28035 Madrid, Spain; (D.P.); (M.R.)
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Mikahela A. López-Morales
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.A.-V.); (M.A.L.-M.); (M.C.-R.)
- Departamento de Fisioterapia, Universidad de Valencia, 46010 Valencia, Spain
| | - Eva Tamayo-Torres
- Departamento de Fisiología, Universidad de Valencia, 46010 Valencia, Spain;
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.A.-V.); (M.A.L.-M.); (M.C.-R.)
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, 46100 Valencia, Spain
| | - David Piñeiro
- AptaTargets S.L., 28035 Madrid, Spain; (D.P.); (M.R.)
| | - Marc Ribó
- AptaTargets S.L., 28035 Madrid, Spain; (D.P.); (M.R.)
- Unidad de Ictus, Departamento de Neurología, Hospital Vall d’Hebron, 08035 Barcelona, Spain
| | - Juan B. Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (A.A.-V.); (M.A.L.-M.); (M.C.-R.)
- Departamento de Fisiología, Universidad de Valencia, 46010 Valencia, Spain;
| |
Collapse
|
31
|
Weber K, Domènech A, Kegler K, Kreutzer R, Mayoral FJ, Okazaki Y, Ortega P, Polledo L, Razinger T, Richard OK, Sanchez R, Warfving N, Vallejo R, de Miguel R. Onset and progression of postmortem histological changes in the central nervous system of RccHan ™: WIST rats. Front Vet Sci 2024; 11:1378609. [PMID: 38835889 PMCID: PMC11149423 DOI: 10.3389/fvets.2024.1378609] [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: 01/29/2024] [Accepted: 03/22/2024] [Indexed: 06/06/2024] Open
Abstract
Death initiates a cascade of physiological and biochemical alterations in organs and tissues, resulting in microscopic changes that challenge the histopathological evaluation. Moreover, the brain is particularly susceptible to artifacts owing to its unique composition and its location within the cranial vault. The aim of this study was to compile and illustrate the microscopic changes in the central nervous system (CNS) of rats subjected to delayed postmortem fixation. It also scrutinizes the influence of exsanguination and cooling methods on the initiation and progression of these alterations. Twenty-four Wistar Han outbred rats (RccHan™: WIST) were sacrificed and stored either at room temperature (18-22°C) or under refrigeration (2-4°C). Necropsies were conducted at different time points postmortem (i.e., 0.5 h, 1 h, 4 h, 8 h, 12 h, 24 h, 36 h, 48 h, 7 days and 14 days). Brain sections underwent simultaneous digital evaluation by 14 pathologists until a consensus was reached on terminology, key findings, and intensity levels. Microscopic observations varied among cell types. Glial cells were similarly affected throughout the CNS and showed pericellular halo, chromatin condensation and nuclear shrinkage. Neurons showed two types of postmortem changes as most of them showed progressive shrinkage, cytoplasmic dissolution and karyorrhexis whereas others acquired a dark-neuron-like appearance. Neuronal changes showed marked differences among neuroanatomical locations. Additional postmortem changes encompassed: granulation and microcavitation in neuropil and white matter; retraction spaces; detachment of ependyma, choroid plexus, and leptomeninges. Severity of findings after 48 h at room temperature was higher than after seven days under refrigeration and similar to or slightly lower than after 14 days under refrigeration. No clear differences were observed related to the sex or weight of the animals or their exsanguination status. This work elucidates the onset and progression of autolytic changes in the brains of Wistar Han rats, offering insights to accurately identify and enhance the histopathological evaluation.
Collapse
|
32
|
König A, Kat CJ, Ganswindt A, Keough N, Oberholzer HM, van Rensburg IJ, Mavunganidze R, Myburgh J. Feasibility of a Sprague???Dawley Rat Model for Investigating the Effects of Seated Whole-body Vibration. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2024; 63:316-324. [PMID: 38508696 PMCID: PMC11193422 DOI: 10.30802/aalas-jaalas-23-000105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/28/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024]
Abstract
Vehicular whole-body vibration (WBV) can have long-term adverse effects on human quality of life. Animal models can be used to study pathophysiologic effects of vibration. The goal of this study was to assess animal cooperation and well-being to determine the feasibility of a novel seated rat model for investigating the effects of WBV on biologic systems. Twenty-four male Sprague???Dawley rats were used. The experiment consisted of an acclimation phase, 2 training phases (TrP1 and TrP2), and a testing phase (TeP), including weekly radiographic imaging. During acclimation, rats were housed in pairs in standard cages without vibration. First, experimental (EG; n = 18) and control group 1 (C1; n = 3) rats were placed in a vibration apparatus without vibration, with increasing duration over 5 d during TrP1. EG rats were exposed to vertical random WBV that was increased in magnitude over 5 d during TrP2 until reaching the vibration signal used during TeP (15min, 0.7m??s-2 root mean square, unweighted). C1 rats were placed in the vibration apparatus but received no vibration during any phase. Control group 2 (C2; n = 3) rats remained in the home cages. Cooperation was evaluated with regard to rat-apparatus interactions and position compliance. Behavior, weight, and fecal glucocorticoid metabolite concentrations (fGCM) were used to evaluate animal well-being. We observed good cooperation and no behavioral patterns or weight loss between phases, indicating little or no animal stress. The differences in fGCM concentration between groups indicated that the EG rats had lower stress levels than the control rats in all phases except TrP1. Thus, this model elicited little or no stress in the conscious, unrestrained, seated rats.
Collapse
Affiliation(s)
- Anya König
- Department of Anatomy, University of Pretoria, Pretoria, South Africa
| | - Cor-Jacques Kat
- Faculty of Engineering, Built Environment and Information Technology, University of Pretoria, Pretoria, South Africa
| | - Andre Ganswindt
- Department of Zoology and Entomology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Natalie Keough
- Department of Anatomy, University of Pretoria, Pretoria, South Africa
- Clinical Anatomy and Imaging, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, UAE; and
| | | | - Ilse Janse van Rensburg
- Onderstepoort Veterinary Animal Research Unit (OVARU), University of Pretoria, Pretoria, South Africa
| | - Richard Mavunganidze
- Onderstepoort Veterinary Animal Research Unit (OVARU), University of Pretoria, Pretoria, South Africa
| | - Jolandie Myburgh
- Department of Anatomy, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
33
|
Amr M, Farid A. Impact of cow, buffalo, goat or camel milk consumption on oxidative stress, inflammation and immune response post weaning time. Sci Rep 2024; 14:9967. [PMID: 38693190 PMCID: PMC11063178 DOI: 10.1038/s41598-024-59959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Milk is a whitish liquid that is secreted from mammary glands; and considered as the primary source of nutrition for newborns since they are not able to digest solid food. However, it contains primary nutrients, as well as growth and immune factors. Early weaning is a critical issue that face women and their babies in developing countries. To avoid infant malnutrition, they tend to use other milk types instead of baby formula. Therefore, the present study aimed to evaluate the impact of cow, buffalo, goat or camel milk consumption on oxidative stress, inflammation and immune response in male and female Sprague Dawley rats post weaning time. The amino acids, fatty acids, minerals and vitamins in the tested milk types were evaluated. Animals were divided into 5 groups (control, cow, buffalo, goat and camel milk administrated groups) (10 rats/group); each animal was administrated by 3.4 ml/day. Rats were administered with milk for 6 weeks; at the end of the 5th week, five animals of each group were isolated and the remaining five animals were immunized with sheep red blood cells (SRBCs) and kept for another week to mount immune response. The effect of different milk types on rats' immune response towards SRBCs was evaluated through pro-inflammatory cytokines, antioxidants, ESR and CRP measurement; together, with the histopathological examination of spleen samples and hemagglutination assay. Camel milk consumption reduced oxidative stress and inflammation in spleen that resulted from SRBCs immunization; in addition to, B cell stimulation that was apparent from the high level of anti-SRBCs antibodies. Camel milk is recommended for newborn consumption, due to its high-water content, unsaturated fatty acids, and vitamin C, as well as low lactose and fat content.
Collapse
Affiliation(s)
- Maryam Amr
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt.
| |
Collapse
|
34
|
Cirilo MAS, Santos VBS, Lima NKS, Muzi-Filho H, Paixão ADO, Vieyra A, Vieira LD. Reactive oxygen species impair Na+ transport and renal components of the renin-angiotensin-aldosterone system after paraquat poisoning. AN ACAD BRAS CIENC 2024; 96:e20230971. [PMID: 38597493 DOI: 10.1590/0001-3765202420230971] [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: 08/28/2023] [Accepted: 11/14/2023] [Indexed: 04/11/2024] Open
Abstract
Paraquat (1,1'-dimethyl-4,4'-bipyridyl dichloride) is an herbicide widely used worldwide and officially banned in Brazil in 2020. Kidney lesions frequently occur, leading to acute kidney injury (AKI) due to exacerbated reactive O2 species (ROS) production. However, the consequences of ROS exposure on ionic transport and the regulator local renin-angiotensin-aldosterone system (RAAS) still need to be elucidated at a molecular level. This study evaluated how ROS acutely influences Na+-transporting ATPases and the renal RAAS. Adult male Wistar rats received paraquat (20 mg/kg; ip). After 24 h, we observed body weight loss and elevation of urinary flow and serum creatinine. In the renal cortex, paraquat increased ROS levels, NADPH oxidase and (Na++K+)ATPase activities, angiotensin II-type 1 receptors, tumor necrosis factor-α (TNF-α), and interleukin-6. In the medulla, paraquat increased ROS levels and NADPH oxidase activity but inhibited (Na++K+)ATPase. Paraquat induced opposite effects on the ouabain-resistant Na+-ATPase in the cortex (decrease) and medulla (increase). These alterations, except for increased serum creatinine and renal levels of TNF-α and interleukin-6, were prevented by 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (tempol; 1 mmol/L in drinking water), a stable antioxidant. In summary, after paraquat poisoning, ROS production culminated with impaired medullary function, urinary fluid loss, and disruption of Na+-transporting ATPases and angiotensin II signaling.
Collapse
Affiliation(s)
- Marry A S Cirilo
- Federal University of Pernambuco, Department of Physiology and Pharmacology, Professor Moraes Rego Ave., University City, 50670-901 Recife, PE, Brazil
| | - Valéria B S Santos
- Federal University of Pernambuco, Department of Physiology and Pharmacology, Professor Moraes Rego Ave., University City, 50670-901 Recife, PE, Brazil
| | - Natália K S Lima
- Federal University of Pernambuco, Department of Physiology and Pharmacology, Professor Moraes Rego Ave., University City, 50670-901 Recife, PE, Brazil
| | - Humberto Muzi-Filho
- Federal University of Rio de Janeiro, Center for Research in Precision Medicine, First Floor, Carlos Chagas Filho Institute of Biophysics, Carlos Chagas Filho Ave., University City, 21941-904 Rio de Janeiro, RJ, Brazil
- Federal University of Rio de Janeiro, National Center for Structural Biology and Bioimaging/CENABIO, 373 Carlos Chagas Filho Ave., University City, 21941-902 Rio de Janeiro, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine-REGENERA, 373 Carlos Chagas Filho Ave., University City, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Ana D O Paixão
- Federal University of Pernambuco, Department of Physiology and Pharmacology, Professor Moraes Rego Ave., University City, 50670-901 Recife, PE, Brazil
| | - Adalberto Vieyra
- Federal University of Rio de Janeiro, Center for Research in Precision Medicine, First Floor, Carlos Chagas Filho Institute of Biophysics, Carlos Chagas Filho Ave., University City, 21941-904 Rio de Janeiro, RJ, Brazil
- Federal University of Rio de Janeiro, National Center for Structural Biology and Bioimaging/CENABIO, 373 Carlos Chagas Filho Ave., University City, 21941-902 Rio de Janeiro, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine-REGENERA, 373 Carlos Chagas Filho Ave., University City, 21941-902 Rio de Janeiro, RJ, Brazil
- Grande Rio University, 1160 Professor José de Souza Herdy Street, Building C, Second Floor, 25071-202 Duque de Caxias, RJ, Brazil
| | - Leucio D Vieira
- Federal University of Pernambuco, Department of Physiology and Pharmacology, Professor Moraes Rego Ave., University City, 50670-901 Recife, PE, Brazil
- Federal University of Rio de Janeiro, National Center for Structural Biology and Bioimaging/CENABIO, 373 Carlos Chagas Filho Ave., University City, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
35
|
Zhang W, Zhang L, Liang W, Wang H, Hu F. Neurodevelopment effects of early life bisphenol-A exposure on visual memory: Insights into recovery dynamics. Toxicology 2024; 502:153718. [PMID: 38160929 DOI: 10.1016/j.tox.2023.153718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Bisphenol A (BPA), a ubiquitous endocrine disruptor, is implicated in the cognitive deficits observed in both children and animals. Especially, BPA-induced spatial memory deterioration during the whole development phase of rodents has been well delineated. However, whether BPA exposure on the different development phases exerts similar effects on the prefrontal cortex (PFC) dependent visual memory is still elusive. Here, we chose two exposure windows, the whole gestation and lactation phases (E0∼P21) and the whole juvenile and adolescent phases (P22∼P60), for exposing rats to BPA. The visual memory of those rats was accessed by object recognition testing in the open field after BPA exposure and a constant recovery interval. The results revealed a substantial decline of visual memory under both exposure conditions, accompanied by an increase in anxiety-like behavior in BPA-exposed rats. Notably, after a 20-day recovery period, those behavioral changes induced by BPA exposure during P22∼60, not E0∼P21, were reversed compared to the control rats. According to morphological analysis of those rats after recovery, we found that the spine density of pyramidal neurons in the PFC were significant decreased in rats with BPA exposure during E0∼P21 and there was no difference between rats with or without BPA exposure during P22∼P60. Additionally, a similar change trend in excitatory receptors expression was observed under both exposure conditions. After an additional 20 days of recovery, the behavioral changes in rats with perinatal BPA exposure reverted to the normal status. Our present findings illuminate the dynamic effects of BPA on PFC-dependent functions across two crucial early developmental stages of life.
Collapse
Affiliation(s)
- Wentai Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Linke Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Weifeng Liang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Huan Wang
- School of Life Science, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Fan Hu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China.
| |
Collapse
|
36
|
Belbis MD, Yap Z, Hobart SE, Ferguson SK, Hirai DM. Effects of acute phosphodiesterase type 5 inhibition on skeletal muscle interstitial PO 2 during contractions and recovery. Nitric Oxide 2024; 142:16-25. [PMID: 37979932 DOI: 10.1016/j.niox.2023.11.004] [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: 07/10/2023] [Revised: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 11/20/2023]
Abstract
The oxygen partial pressure within the interstitial space (PO2is; mmHg) provides the driving force for oxygen diffusion into the myocyte thereby supporting oxidative phosphorylation. We tested the hypothesis that potentiation of the nitric oxide pathway with sildenafil (phosphodiesterase type 5 inhibitor) would enhance PO2is during muscle metabolic transitions, thereby slowing PO2is on- and accelerating PO2is off-kinetics. The rat spinotrapezius muscle (n = 17) was exposed for PO2is measurements via phosphorescence quenching under control (CON), low-dose sildenafil (1 mg/kg i.a., SIL1) and high-dose sildenafil (7 mg/kg i.a., SIL7). Data were collected at rest and during submaximal twitch contractions (1 Hz, 4-6 V, 3 min) and recovery (3 min). Mean arterial blood pressure (MAP; mmHg) was reduced with both SIL1 (pre:132 ± 5; post:99 ± 5) and SIL7 (pre:111 ± 6; post:99 ± 4) (p < 0.05). SIL7 elevated resting PO2is (18.4 ± 1.1) relative to both CON (15.7 ± 0.7) and SIL1 (15.2 ± 0.7) (p < 0.05). In addition, SIL7 increased end-recovery PO2is (17.7 ± 1.6) compared to CON (12.8 ± 0.9) and SIL1 (13.4 ± 0.8) (p < 0.05). The overall PO2is response during recovery (i.e., area under the PO2is curve) was greater in SIL7 (4107 ± 444) compared to CON (3493 ± 222) and SIL1 (3114 ± 205 mmHg s) (p < 0.05). Contrary to our hypothesis, there was no impact of acute SIL (1 or 7 mg/kg) on the speed of the PO2is response during contractions or recovery (p > 0.05). However, sildenafil lowered MAP and improved skeletal muscle interstitial oxygenation in healthy rats. Specifically, SIL7 enhanced PO2is at rest and during recovery from submaximal muscle contractions. Potentiation of the nitric oxide pathway with sildenafil enhances microvascular blood-myocyte O2 transport and is expected to improve repeated bouts of contractile activity.
Collapse
Affiliation(s)
- Michael D Belbis
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA; Department of Exercise Science, Aurora University, Aurora, IL, USA
| | - Zhen Yap
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Sara E Hobart
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Scott K Ferguson
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL, USA
| | - Daniel M Hirai
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
37
|
Kalyanaraman H, Casteel DE, Cabriales JA, Tat J, Zhuang S, Chan A, Dretchen KL, Boss GR, Pilz RB. The Antioxidant/Nitric Oxide-Quenching Agent Cobinamide Prevents Aortic Disease in a Mouse Model of Marfan Syndrome. JACC Basic Transl Sci 2024; 9:46-62. [PMID: 38362350 PMCID: PMC10864892 DOI: 10.1016/j.jacbts.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 02/17/2024]
Abstract
Major pathologic changes in the proximal aorta underlie the life-threatening aortic aneurysms and dissections in Marfan Syndrome; current treatments delay aneurysm development without addressing the primary pathology. Because excess oxidative stress and nitric oxide/protein kinase G signaling likely contribute to the aortopathy, we hypothesized that cobinamide, a strong antioxidant that can attenuate nitric oxide signaling, could be uniquely suited to prevent aortic disease. In a well-characterized mouse model of Marfan Syndrome, cobinamide dramatically reduced elastin breaks, prevented excess collagen deposition and smooth muscle cell apoptosis, and blocked DNA, lipid, and protein oxidation and excess nitric oxide/protein kinase G signaling in the ascending aorta. Consistent with preventing pathologic changes, cobinamide diminished aortic root dilation without affecting blood pressure. Cobinamide exhibited excellent safety and pharmacokinetic profiles indicating it could be a practical treatment. We conclude that cobinamide deserves further study as a disease-modifying treatment of Marfan Syndrome.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Darren E. Casteel
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Justin A. Cabriales
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - John Tat
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Adriano Chan
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | | | - Gerry R. Boss
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Renate B. Pilz
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| |
Collapse
|
38
|
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
|
39
|
Adedeji AO, Zhong F, Corpuz J, Hu F, Zhao X, Sangaraju D, Ruff CF, Dybdal N. Comparative Impact of Various Fasting Periods on the Welfare of Sprague-Dawley Rats With or Without Supplementation. Toxicol Pathol 2024; 52:21-34. [PMID: 38379371 DOI: 10.1177/01926233241230536] [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: 02/22/2024]
Abstract
In nonclinical toxicology studies, lab animals are fasted typically overnight, to reduce variability in some clinical pathology parameters. However, fasting adds undue stress, and this is particularly concerning in rodents given their fast metabolic rates. Furthermore, as rodents are nocturnal animals, an overnight fasting may cause a protracted negative metabolic state even when the fasting has technically ended, given their minimal activity and food consumption during the day. Therefore, to evaluate the impacts of different fasting durations (±DietGel supplementation) on rats' welfare, we assessed the traditional and ancillary clinical pathology parameters in Sprague-Dawley rats, along with body weight, organ weight, and histopathology. Although most endpoints were comparable between the different fasting durations (±DietGel supplementation), the long fasting times (≥8 hr) without DietGel supplementation caused significant decreases in body weight, liver weight, liver glycogen content, serum glucose, triglyceride, and creatinine concentrations-all findings suggestive of a negative energy balance that could impact animal welfare and consequently, data quality; while the short fasting time (4 hr) and DietGel supplementation were associated with higher triglycerides variability. Hence, we propose that short fasting time should be adequate for most toxicology studies in rats, and long fasting times should only be accommodated with scientific justification.
Collapse
Affiliation(s)
- Adeyemi O Adedeji
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Fiona Zhong
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Janice Corpuz
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Fangyao Hu
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Xiaofeng Zhao
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Dewakar Sangaraju
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Catherine F Ruff
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Noel Dybdal
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| |
Collapse
|
40
|
Poulios C, Karagkiozaki V, Kapoukranidou D, Chakim Z, Zarampoukas T, Foroglou N, Logothetidis S. Bringing pathology to nanomedicine: a comparison of in vivo toxicity of polymeric nanoparticle carriers with and without chitosan coating. Virchows Arch 2023; 483:775-786. [PMID: 37402995 DOI: 10.1007/s00428-023-03581-y] [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: 03/18/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 07/06/2023]
Abstract
Over the last years, there has been an increasing number of proposals for the use of nanomaterials in medicine. The safety of novel technologies must be verified, prior to their clinical application. Pathology has much to contribute towards this end. In this study, we compared the in vivo toxicity effects of poly- (lactic-co-glycolic acid) nanoparticles with and without chitosan shell. Both nanoparticle types were loaded with curcumin. The nanoparticles were assessed in vitro for potential cytotoxicity with cell viability studies. For the in vivo test, 36 adult Wistar rats were used, four of which were the control group. The remaining 32 were divided into 2 groups, each of which was administered differentially coated drug carriers: (A) nanoparticles without chitosan coating and (B) nanoparticles with chitosan coating. For both groups, the subcutaneous route was used for administration. Each group was further divided into 2 sub-groups of 8 animals each. The animals of the first sub-groups were sacrificed 24 h after the injection and those of the second on the 7th day. The control group was also divided into 2 subgroups of 2 animals each. At the appointed post-administrative date, the rats were sacrificed, and specimens from the brain, liver, kidneys, heart, stomach, lungs, and from the skin at the injection site were collected and studied histopathologically. The evaluation of both in vitro and in vivo testing shows that nanoparticles with chitosan have significantly less, if any, toxic effects compared to those without chitosan.
Collapse
Affiliation(s)
- Christos Poulios
- Department of Pathology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
- European Society of Pathology, Brussels, Belgium.
| | - Varvara Karagkiozaki
- Laboratory of Thin Films, Nanobiomaterials-Nanosystems and Nanometrology, Faculty of Physics, Aristotle University of Thessaloniki, Thessaloniki, Greece
- BL NanoBiomed, Thessaloniki, Greece
| | - Dorothea Kapoukranidou
- Department of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Zena Chakim
- Laboratory of Thin Films, Nanobiomaterials-Nanosystems and Nanometrology, Faculty of Physics, Aristotle University of Thessaloniki, Thessaloniki, Greece
- BL NanoBiomed, Thessaloniki, Greece
| | - Thomas Zarampoukas
- Department of Pathology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Foroglou
- Department of Neurosurgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios Logothetidis
- Laboratory of Thin Films, Nanobiomaterials-Nanosystems and Nanometrology, Faculty of Physics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
41
|
Marzi SJ, Schilder BM, Nott A, Frigerio CS, Willaime-Morawek S, Bucholc M, Hanger DP, James C, Lewis PA, Lourida I, Noble W, Rodriguez-Algarra F, Sharif JA, Tsalenchuk M, Winchester LM, Yaman Ü, Yao Z, Ranson JM, Llewellyn DJ. Artificial intelligence for neurodegenerative experimental models. Alzheimers Dement 2023; 19:5970-5987. [PMID: 37768001 DOI: 10.1002/alz.13479] [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: 04/17/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023]
Abstract
INTRODUCTION Experimental models are essential tools in neurodegenerative disease research. However, the translation of insights and drugs discovered in model systems has proven immensely challenging, marred by high failure rates in human clinical trials. METHODS Here we review the application of artificial intelligence (AI) and machine learning (ML) in experimental medicine for dementia research. RESULTS Considering the specific challenges of reproducibility and translation between other species or model systems and human biology in preclinical dementia research, we highlight best practices and resources that can be leveraged to quantify and evaluate translatability. We then evaluate how AI and ML approaches could be applied to enhance both cross-model reproducibility and translation to human biology, while sustaining biological interpretability. DISCUSSION AI and ML approaches in experimental medicine remain in their infancy. However, they have great potential to strengthen preclinical research and translation if based upon adequate, robust, and reproducible experimental data. HIGHLIGHTS There are increasing applications of AI in experimental medicine. We identified issues in reproducibility, cross-species translation, and data curation in the field. Our review highlights data resources and AI approaches as solutions. Multi-omics analysis with AI offers exciting future possibilities in drug discovery.
Collapse
Affiliation(s)
- Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Brian M Schilder
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | | | - Magda Bucholc
- School of Computing, Engineering & Intelligent Systems, Ulster University, Derry, UK
| | - Diane P Hanger
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Patrick A Lewis
- Royal Veterinary College, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Wendy Noble
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Jalil-Ahmad Sharif
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Maria Tsalenchuk
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Ümran Yaman
- UK Dementia Research Institute at UCL, London, UK
| | | | | | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- Alan Turing Institute, London, UK
| |
Collapse
|
42
|
Jaffer H, Andrabi SS, Petro M, Kuang Y, Steinmetz MP, Labhasetwar V. Catalytic antioxidant nanoparticles mitigate secondary injury progression and promote functional recovery in spinal cord injury model. J Control Release 2023; 364:109-123. [PMID: 37866402 PMCID: PMC10842504 DOI: 10.1016/j.jconrel.2023.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Traumatic spinal cord injury exacerbates disability with time due to secondary injury cascade triggered largely by overproduction of reactive oxygen species (ROS) at the lesion site, causing oxidative stress. This study explored nanoparticles containing antioxidant enzymes (antioxidant NPs) to neutralize excess ROS at the lesion site and its impact. When tested in a rat contusion model of spinal cord injury, a single dose of antioxidant NPs, administered intravenously three hours after injury, effectively restored the redox balance at the lesion site, interrupting the secondary injury progression. This led to reduced spinal cord tissue inflammation, apoptosis, cavitation, and inhibition of syringomyelia. Moreover, the treatment reduced scar tissue forming collagen at the lesion site, protected axons from demyelination, and stimulated lesion healing, with further analysis indicating the formation of immature neurons. The ultimate effect of the treatment was improved motor and sensory functions and rapid post-injury weight loss recovery. Histological analysis revealed activated microglia in the spinal cord displaying rod-shaped anti-inflammatory and regenerative phenotype in treated animals, contrasting with amoeboid inflammatory and degenerative phenotype in untreated control. Overall data suggest that restoring redox balance at the lesion site shifts the dynamics in the injured spinal cord microenvironment from degenerative to regenerative, potentially by promoting endogenous repair mechanisms. Antioxidant NPs show promise to be developed as an early therapeutic intervention in stabilizing injured spinal cord for enhanced recovery.
Collapse
Affiliation(s)
- Hayder Jaffer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Syed Suhail Andrabi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marianne Petro
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Youzhi Kuang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael P Steinmetz
- Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Flores-Estrada J, Cano-Martínez A, Vargas-González Á, Castrejón-Téllez V, Cornejo-Garrido J, Martínez-Rosas M, Guarner-Lans V, Rubio-Ruíz ME. Hepatoprotective Mechanisms Induced by Spinach Methanolic Extract in Rats with Hyperglycemia-An Immunohistochemical Analysis. Antioxidants (Basel) 2023; 12:2013. [PMID: 38001866 PMCID: PMC10669258 DOI: 10.3390/antiox12112013] [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: 10/23/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Spinach methanolic extract (SME) has a hepatoprotective effect due to its polyphenolic antioxidants; however, its action in parenchymal (PQ) and non-parenchymal (nPQ) cells remains unknown. This study investigates the hepatoprotective effect of SME on streptozotocin-induced hyperglycemic rats (STZ), focusing on immunohistochemical analyses. Methods: The extract was prepared, and the total polyphenols and antioxidant activity were quantified. Adult male Wistar rats were divided into four groups (n = 8): normoglycemic rats (NG), STZ-induced hyperglycemic (STZ), STZ treated with 400 mg/kg SME (STZ-SME), and NG treated with SME (SME) for 12 weeks. Serum liver transaminases and lipid peroxidation levels in tissue were determined. The distribution pattern and relative levels of markers related to oxidative stress [reactive oxygen species (ROS), superoxide dismutase-1, catalase, and glutathione peroxidase-1], of cytoprotective molecules [nuclear NRF2 and heme oxygenase-1 (HO-1)], of inflammatory mediators [nuclear NF-κB, TNF-α], proliferation (PCNA), and of fibrogenesis markers [TGF-β, Smad2/3, MMP-9, and TIMP1] were evaluated. Results: SME had antioxidant capacity, and it lowered serum transaminase levels in STZ-SME compared to STZ. It reduced NOX4 staining, and lipid peroxidation levels were related to low formation of ROS. In STZ-SME, the immunostaining for antioxidant enzymes increased in nPQ cells compared to STZ. However, enzymes were also localized in extra and intracellular vesicles in STZ. Nuclear NRF2 staining and HO-1 expression in PQ and nPQ were higher in STZ-SME than in STZ. Inflammatory factors were decreased in STZ-SME and were related to the percentage decrease in NF-κB nuclear staining in nPQ cells. Similarly, TGF-β (in the sinusoids) and MMP-9 (in nPQ) were increased in the STZ-SME group compared to the other groups; however, staining for CTGF, TIMP1, and Smad2/3 was lower. Conclusions: SME treatment in hyperglycemic rats induced by STZ may have hepatoprotective properties due to its scavenger capacity and the regulation of differential expression of antioxidant enzymes between the PQ and nPQ cells, reducing inflammatory and fibrogenic biomarkers in liver tissue.
Collapse
Affiliation(s)
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| | - Álvaro Vargas-González
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| | - Vicente Castrejón-Téllez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| | - Jorge Cornejo-Garrido
- Laboratorio de Biología Celular y Productos Naturales, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional, Mexico City 07320, Mexico;
| | - Martín Martínez-Rosas
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| | - Verónica Guarner-Lans
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| | - María Esther Rubio-Ruíz
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.); (Á.V.-G.); (V.C.-T.); (M.M.-R.); (V.G.-L.); (M.E.R.-R.)
| |
Collapse
|
44
|
Vasanthi SS, Massey N, Nair SN, Mochel JP, Showman L, Thippeswamy T. Exploring the benefits of in-diet versus repeated oral dosing of saracatinib (AZD0530) in chronic studies: insights into pharmacokinetics and animal welfare. Front Vet Sci 2023; 10:1297221. [PMID: 38026620 PMCID: PMC10666625 DOI: 10.3389/fvets.2023.1297221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Saracatinib/AZD0530 (SAR), a Src tyrosine kinase inhibitor, mitigates seizure-induced brain pathology in epilepsy models upon repeated oral dosing. However, repeated dosing is stressful and can be challenging in some seizing animals. To overcome this issue, we have incorporated SAR-in-Diet and compared serum pharmacokinetics (PK) and brain concentrations with conventional repeated oral dosing. Saracatinib in solution or in-diet was stable at room temperature for >4 weeks (97 ± 1.56%). Adult Sprague Dawley rats on SAR-in-Diet consumed ~1.7 g/day less compared to regular diet (16.82 ± 0.6 vs. 18.50 ± 0.5 g/day), but the weight gain/day was unaffected (2.63 ± 0.5 g/day vs. 2.83 ± 0.2 g/day). Importantly, we achieved the anticipated SAR dose range from 2.5-18.7 mg/kg of rat in response to varying concentrations of SAR-in-Diet from 54 to 260 ppm of feed, respectively. There was a strong and significant correlation between SAR-in-Diet dose (mg/kg) and serum saracatinib concentrations (ng/ml). Serum concentrations also did not vary significantly between SAR-in-Diet and repeated oral dosing. The hippocampal saracatinib concentrations derived from SAR-in-Diet treatment were higher than those derived after repeated oral dosing (day 3, 546.8 ± 219.7 ng/g vs. 238.6 ± 143 ng/g; day 7, 300.7 ± 43.4 ng/g vs. 271.1 ± 62.33 ng/g). Saracatinib stability at room temperature and high serum and hippocampal concentrations in animals fed on SAR-in-Diet are useful to titer the saracatinib dose for future animal disease models. Overall, test drugs in the diet is an experimental approach that addresses issues related to handling stress-induced variables in animal experiments.
Collapse
Affiliation(s)
- Suraj S. Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Nyzil Massey
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Suresh N. Nair
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Kerala Veterinary and Animal Sciences University, Thrissur, India
| | - Jonathan P. Mochel
- Precision One Health, Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Lucas Showman
- W.M. Keck Metabolomics Research Laboratory, Iowa State University, Ames, IA, United States
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
45
|
Amr M, Mohie-Eldinn M, Farid A. Evaluation of buffalo, cow, goat and camel milk consumption on multiple health outcomes in male and female Sprague Dawley rats. Int Dairy J 2023; 146:105760. [DOI: 10.1016/j.idairyj.2023.105760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
|
46
|
Aroniadou-Anderjaska V, Figueiredo TH, de Araujo Furtado M, Pidoplichko VI, Braga MFM. Mechanisms of Organophosphate Toxicity and the Role of Acetylcholinesterase Inhibition. TOXICS 2023; 11:866. [PMID: 37888716 PMCID: PMC10611379 DOI: 10.3390/toxics11100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Organophosphorus compounds (OPs) have applications in agriculture (e.g., pesticides), industry (e.g., flame retardants), and chemical warfare (nerve agents). In high doses or chronic exposure, they can be toxic or lethal. The primary mechanism, common among all OPs, that initiates their toxic effects is the inhibition of acetylcholinesterase. In acute OP exposure, the subsequent surge of acetylcholine in cholinergic synapses causes a peripheral cholinergic crisis and status epilepticus (SE), either of which can lead to death. If death is averted without effective seizure control, long-term brain damage ensues. This review describes the mechanisms by which elevated acetylcholine can cause respiratory failure and trigger SE; the role of the amygdala in seizure initiation; the role of M1 muscarinic receptors in the early stages of SE; the neurotoxic pathways activated by SE (excitotoxicity/Ca++ overload/oxidative stress, neuroinflammation); and neurotoxic mechanisms linked to low-dose, chronic exposure (Ca++ dyshomeostasis/oxidative stress, inflammation), which do not depend on SE and do not necessarily involve acetylcholinesterase inhibition. The evidence so far indicates that brain damage from acute OP exposure is a direct result of SE, while the neurotoxic mechanisms activated by low-dose chronic exposure are independent of SE and may not be associated with acetylcholinesterase inhibition.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (V.A.-A.); (V.I.P.)
- Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Taiza H. Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (V.A.-A.); (V.I.P.)
| | - Marcio de Araujo Furtado
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (V.A.-A.); (V.I.P.)
| | - Volodymyr I. Pidoplichko
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (V.A.-A.); (V.I.P.)
| | - Maria F. M. Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (V.A.-A.); (V.I.P.)
- Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
47
|
Kumar R, Rizvi SI. Vitamin C Improves Inflammatory-related Redox Status in Hyperlipidemic Rats. Indian J Clin Biochem 2023; 38:512-518. [PMID: 37746546 PMCID: PMC10516823 DOI: 10.1007/s12291-022-01070-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022]
Abstract
Excessive dietary fat is mainly responsible for metabolic diseases including atherosclerosis and cardiovascular disease. We have evaluated the role of Vitamin C in an experimental hyperlipidemic model of rats (male Wistar rat 12-16 months). The hyperlipidemic model of the rat was created by treatment with an atherogenic suspension: cholesterol, cholic acid, and coconut oil, for 30 days once daily, and supplemented with Vitamin C (Ascorbic acid) doses of 0.5 g/kg body weight (orally) for the 30 days once daily. Bodyweight, fasting glucose, triglyceride, cholesterol, ROS (Reactive oxygen species), MDA (Malondialdehyde), FRAP (Ferric reducing the ability of plasma), GSH (Reduced glutathione), PCO (Protein carbonyl), PON-1(Paraoxonase-1), AGE (Advanced glycation end product), PMRS (Plasma membrane reduced system), and inflammatory cytokines (TNF-α and IL-6) were estimated in blood and plasma. Our result shows that oxidative stress, and inflammatory markers, were increased in the HFD-treated group of rats. Vitamin C supplementation protected against lipidemic and, oxidative stress. We conclude that Vitamin C may be useful in maintaining cellular redox balance and protecting against lipidemic stress.
Collapse
Affiliation(s)
- Raushan Kumar
- Department of Biochemistry, University of Allahabad, 211002 Allahabad, India
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, 211002 Allahabad, India
| |
Collapse
|
48
|
McNerney MW, Gurkoff GG, Beard C, Berryhill ME. The Rehabilitation Potential of Neurostimulation for Mild Traumatic Brain Injury in Animal and Human Studies. Brain Sci 2023; 13:1402. [PMID: 37891771 PMCID: PMC10605899 DOI: 10.3390/brainsci13101402] [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: 08/14/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Neurostimulation carries high therapeutic potential, accompanied by an excellent safety profile. In this review, we argue that an arena in which these tools could provide breakthrough benefits is traumatic brain injury (TBI). TBI is a major health problem worldwide, with the majority of cases identified as mild TBI (mTBI). MTBI is of concern because it is a modifiable risk factor for dementia. A major challenge in studying mTBI is its inherent heterogeneity across a large feature space (e.g., etiology, age of injury, sex, treatment, initial health status, etc.). Parallel lines of research in human and rodent mTBI can be collated to take advantage of the full suite of neuroscience tools, from neuroimaging (electroencephalography: EEG; functional magnetic resonance imaging: fMRI; diffusion tensor imaging: DTI) to biochemical assays. Despite these attractive components and the need for effective treatments, there are at least two major challenges to implementation. First, there is insufficient understanding of how neurostimulation alters neural mechanisms. Second, there is insufficient understanding of how mTBI alters neural function. The goal of this review is to assemble interrelated but disparate areas of research to identify important gaps in knowledge impeding the implementation of neurostimulation.
Collapse
Affiliation(s)
- M. Windy McNerney
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA; (M.W.M.); (C.B.)
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gene G. Gurkoff
- Department of Neurological Surgery, and Center for Neuroscience, University of California, Davis, Sacramento, CA 95817, USA;
- Department of Veterans Affairs, VA Northern California Health Care System, Martinez, CA 94553, USA
| | - Charlotte Beard
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA; (M.W.M.); (C.B.)
- Program in Neuroscience and Behavioral Biology, Emory University, Atlanta, GA 30322, USA
| | - Marian E. Berryhill
- Programs in Cognitive and Brain Sciences, and Integrative Neuroscience, Department of Psychology, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
49
|
Garcia-Alonso I, Velasco-Oraa X, Cearra I, Iturrizaga Correcher S, Mar Medina C, Alonso-Varona A, García Ruiz de Gordejuela A, Ruiz-Montesinos I, Herrero de la Parte B. Prophylactic Treatment of Intestinal Ischemia-Reperfusion Injury Reduces Mucosal Damage and Improves Intestinal Absorption. J Inflamm Res 2023; 16:4141-4152. [PMID: 37750172 PMCID: PMC10518153 DOI: 10.2147/jir.s426396] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/02/2023] [Indexed: 09/27/2023] Open
Abstract
Purpose Intestinal ischemia-reperfusion injury (i-IRI) involves a blood flow interruption in an intestinal segment followed by blood flow restoration. When blood flow is restored, oxidative and inflammatory molecules are distributed throughout the bloodstream, triggering both local and systemic damage. Our goal was to evaluate the potential of three antioxidant and/or anti-inflammatory compounds (curcumin, dexmedetomidine and α-tocopherol) to prevent or reverse local and systemic damage induced by i-IRI. Methods i-IRI was induced by placing a microvascular clip in the superior mesenteric artery of female WAG/RijHsd rats; the clip was removed after 1h and reperfusion was allowed for 4h. Curcumin (200 mg/kg, orally), α-tocopherol (20 mg/kg, i.p.), and dexmedetomidine (5 or 20 µg/kg, s.c.; DEX5 and DEX20, respectively) were administered. Blood and terminal ileum specimens were collected for biochemical and histological determination. Furthermore, D-xylose absorption test was performed to evaluate intestinal absorption; after completing the 1-hour ischemia and 4-hour reperfusion period, 1 mL of aqueous D-xylose solution (0.615 mg/mL) was administered orally, and one hour later, plasma D-xylose levels were quantified. Results The histological injury degree (HID) measured by the Chiu scale was significantly reduced when the treatments were applied (non-treated rats, 2.6 ± 0.75; curcumin, 1.54 ± 0.8; DEX5, 1.47 ± 0.7; DEX20 1.14 ± 0.5; and α-tocopherol, 1.01 ± 0.6); intestinal absorptive capacity also improved in all cases healthy rats (2.06 ± 0.07 µg/mL; non-treated, 1.18 ± 0.07 µg/mL; curcumin 1.76 ± 0.3 µg/mL; DEX5, 2.29 ± 0.2 µg/mL; DEX20, 2.25 ± 0.26 µg/mL; and α-tocopherol 1.66 ± 0.21 µg/mL). However, it failed to reduce liver enzyme levels. Finally, only dexmedetomidine significantly reduced urea and creatinine levels compared to non-treated animals. Conclusion All drugs were effective in reducing HID, although α-tocopherol was effective to a greater extent. Only dexmedetomidine reverted intestinal absorption to normal values of healthy animals.
Collapse
Affiliation(s)
- Ignacio Garcia-Alonso
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Interventional Radiology Research Group, Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
| | - Xabier Velasco-Oraa
- Department of Anaesthesia, Hospital Clínic of Barcelona, Barcelona, 08036, Spain
| | - Iñigo Cearra
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Department of Orthopedics, Basurto University Hospital, Osakidetza Basque Health Service, Bilbao, 48013, Spain
- Regenerative Therapies, Osteoarticular and Tendon Pathology Research Group, Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
| | | | - Carmen Mar Medina
- Department of Clinical Analyses, Galdakao-Usansolo University Hospital, Galdakao, 48960, Spain
| | - Ana Alonso-Varona
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| | - Amador García Ruiz de Gordejuela
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Department of Gastrointestinal Surgery, Donostia University Hospital, Osakidetza Basque Health Service, Donostia, 20014, Spain
| | - Inmaculada Ruiz-Montesinos
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Department of Gastrointestinal Surgery, Donostia University Hospital, Osakidetza Basque Health Service, Donostia, 20014, Spain
| | - Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Interventional Radiology Research Group, Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
| |
Collapse
|
50
|
Lyden PD, Diniz MA, Bosetti F, Lamb J, Nagarkatti KA, Rogatko A, Kim S, Cabeen RP, Koenig JI, Akhter K, Arbab AS, Avery BD, Beatty HE, Bibic A, Cao S, Simoes Braga Boisserand L, Chamorro A, Chauhan A, Diaz-Perez S, Dhandapani K, Dhanesha N, Goh A, Herman AL, Hyder F, Imai T, Johnson CW, Khan MB, Kamat P, Karuppagounder SS, Kumskova M, Mihailovic JM, Mandeville JB, Morais A, Patel RB, Sanganahalli BG, Smith C, Shi Y, Sutariya B, Thedens D, Qin T, Velazquez SE, Aronowski J, Ayata C, Chauhan AK, Leira EC, Hess DC, Koehler RC, McCullough LD, Sansing LH. A multi-laboratory preclinical trial in rodents to assess treatment candidates for acute ischemic stroke. Sci Transl Med 2023; 15:eadg8656. [PMID: 37729432 DOI: 10.1126/scitranslmed.adg8656] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Human diseases may be modeled in animals to allow preclinical assessment of putative new clinical interventions. Recent, highly publicized failures of large clinical trials called into question the rigor, design, and value of preclinical assessment. We established the Stroke Preclinical Assessment Network (SPAN) to design and implement a randomized, controlled, blinded, multi-laboratory trial for the rigorous assessment of candidate stroke treatments combined with intravascular thrombectomy. Efficacy and futility boundaries in a multi-arm multi-stage statistical design aimed to exclude from further study highly effective or futile interventions after each of four sequential stages. Six independent research laboratories performed a standard focal cerebral ischemic insult in five animal models that included equal numbers of males and females: young mice, young rats, aging mice, mice with diet-induced obesity, and spontaneously hypertensive rats. The laboratories adhered to a common protocol and efficiently enrolled 2615 animals with full data completion and comprehensive animal tracking. SPAN successfully implemented treatment masking, randomization, prerandomization inclusion and exclusion criteria, and blinded assessment of outcomes. The SPAN design and infrastructure provide an effective approach that could be used in similar preclinical, multi-laboratory studies in other disease areas and should help improve reproducibility in translational science.
Collapse
Affiliation(s)
- Patrick D Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Márcio A Diniz
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Francesca Bosetti
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Karisma A Nagarkatti
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - André Rogatko
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sungjin Kim
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ryan P Cabeen
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Imaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kazi Akhter
- Department of Radiology, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | - Ali S Arbab
- Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912-0004, USA
| | - Brooklyn D Avery
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | - Hannah E Beatty
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Adnan Bibic
- Department of Radiology, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | - Suyi Cao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | | | - Angel Chamorro
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona 08036, Spain
| | - Anjali Chauhan
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX 77030, USA
| | - Sebastian Diaz-Perez
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Krishnan Dhandapani
- Department Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nirav Dhanesha
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew Goh
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX 77030, USA
| | - Alison L Herman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Takahiko Imai
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Conor W Johnson
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mohammad B Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Pradip Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | | | - Mariia Kumskova
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jelena M Mihailovic
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Joseph B Mandeville
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Andreia Morais
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Rakesh B Patel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | - Cameron Smith
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yanrong Shi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | - Brijesh Sutariya
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel Thedens
- Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Tao Qin
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Sofia E Velazquez
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX 77030, USA
| | - Cenk Ayata
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Enrique C Leira
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218-2625, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX 77030, USA
| | - Lauren H Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|