Product Citations: 80

1 image found

Juvenile mice are susceptible to infarct-induced neurodegeneration that causes delayed cognitive decline in a new model of pediatric ischemic stroke

Preprint on BioRxiv : the Preprint Server for Biology on 16 April 2025 by Mayne, E. W., Vanden, K., et al.

ABSTRACT Over half of pediatric stroke survivors have permanent neurologic and cognitive deficits, and some children develop new or worsening cognitive impairment late after stroke. Their cognitive symptoms are associated with chronic alterations of brain structure in regions distant from the infarct, including in the uninjured hemisphere. However, the mechanisms that cause children’s late cognitive symptoms and disrupted brain growth outside the infarcted tissue remain unknown. We therefore developed and validated a mouse model of pediatric ischemic stroke that causes infarct-induced, delayed cognitive decline accompanied by chronic innate and adaptive neuroinflammation in the infarct and in uninjured subcortical regions that are connected to the infarct. Methods Male and female C57BL/6J mice were randomized to stroke or sham surgery at p28 to model stroke in late childhood and compared to adult 6-month-old mice. We used permanent distal MCAO followed by 60 minutes of hypoxia, an established model for focal ischemia that generates a purely cortical stroke. Mice underwent behavioral testing at 1- and 7-weeks post stroke Barnes Maze protocol adapted for reversal learning. We performed immunostaining to quantify stroke size, atrophy, and innate and adaptive neuroinflammation at 3 days and 7 weeks after surgery. Results At 7 weeks post-stroke, juvenile stroke mice performed significantly worse on reversal learning on Barnes Maze testing than sham mice (n = 7 stroke, 7 sham, p = 0.0265 2-way ANOVA with repeated measures). At 3 days after stroke, juvenile mice had greater innate immune cell activation at subcortical sites of secondary neurodegeneration; however, by 7 weeks after surgery, this relationship was reversed, and adults had greater chronic innate immune cell activation at these uninjured subcortical sites (corpus callosum, corticospinal tract, thalamus). Like adult mice, pediatric mice exhibited chronic innate and adaptive neuroimmunity in the infarct scar; the inflammation in the stroke scar did not differ by age. Conclusions In a model of pediatric ischemic stroke, juvenile mice develop a delayed cognitive deficit that is associated with chronic innate and adaptive neuroinflammation in uninjured subcortical structures that undergo secondary neurodegeneration. Our results suggest that infarct-induced neurodegeneration occurs after stroke in juvenile mice and that juvenile and adult mice have divergent trajectories in the innate immune response to stroke.

  • Cardiovascular biology
  • Neuroscience

Delivering macromolecules across biological barriers such as the blood-brain barrier limits their application in vivo. Previous work has demonstrated that Toxoplasma gondii, a parasite that naturally travels from the human gut to the central nervous system (CNS), can deliver proteins to host cells. Here we engineered T. gondii's endogenous secretion systems, the rhoptries and dense granules, to deliver multiple large (>100 kDa) therapeutic proteins into neurons via translational fusions to toxofilin and GRA16. We demonstrate delivery in cultured cells, brain organoids and in vivo, and probe protein activity using imaging, pull-down assays, scRNA-seq and fluorescent reporters. We demonstrate robust delivery after intraperitoneal administration in mice and characterize 3D distribution throughout the brain. As proof of concept, we demonstrate GRA16-mediated brain delivery of the MeCP2 protein, a putative therapeutic target for Rett syndrome. By characterizing the potential and current limitations of the system, we aim to guide future improvements that will be required for broader application.
© 2024. The Author(s).

  • Mus musculus (House mouse)
  • Neuroscience

Chronic consequences of ischemic stroke: Profiling brain injury and inflammation in a mouse model with reperfusion.

In Physiological Reports on 1 June 2024 by Murphy, D. P., Dickson, D. C., et al.

Stroke is a pervasive and debilitating global health concern, necessitating innovative therapeutic strategies, especially during recovery. While existing literature often focuses on acute interventions, our study addresses the uniqueness of brain tissue during wound healing, emphasizing the chronic phase following the commonly used middle cerebral artery (MCA) occlusion model. Using clinically relevant endpoints in male and female mice such as magnetic resonance imaging (MRI) and plasma neurofilament light (NFL) measurement, along with immunohistochemistry, we describe injury evolution. Our findings document significant alterations in edema, tissue remodeling, and gadolinium leakage through MRI. Plasma NFL concentration remained elevated at 30 days poststroke. Microglia responses are confined to the region adjacent to the injury, rather than continued widespread activation, and boron-dipyrromethene (BODIPY) staining demonstrated the persistent presence of foam cells within the infarct. Additional immunohistochemistry highlighted sustained B and T lymphocyte presence in the poststroke brain. These observations underscore potentially pivotal roles played by chronic inflammation brought on by the lipid-rich brain environment, and chronic blood-brain barrier dysfunction, in the development of secondary neurodegeneration. This study sheds light on the enduring consequences of ischemic stroke in the most used rodent stroke model and provides valuable insights for future research, clinical strategies, and therapeutic development.© 2024 The Author(s). Physiological Reports published by Wiley Periodicals LLC on behalf of The Physiological Society and the American Physiological Society.

  • IHC
  • Mus musculus (House mouse)
  • Cardiovascular biology
  • Immunology and Microbiology

Change within the intratumoral microbiome is a common feature in lung and other cancers and may influence inflammation and immunity in the tumor microenvironment, affecting growth and metastases. We previously characterized the lung cancer microbiome in patients and identified Acidovorax temperans as enriched in tumors. Here, we instilled A. temperans in an animal model driven by mutant K-ras and Tp53. This revealed A. temperans accelerates tumor development and burden through infiltration of proinflammatory cells. Neutrophils exposed to A. temperans displayed a mature, pro-tumorigenic phenotype with increased cytokine signaling, with a global shift away from IL-1β signaling. Neutrophil to monocyte and macrophage signaling upregulated MHC II to activate CD4+ T cells, polarizing them to an IL-17A+ phenotype detectable in CD4+ and γδ populations (T17). These T17 cells shared a common gene expression program predictive of poor survival in human LUAD. These data indicate bacterial exposure promotes tumor growth by modulating inflammation.
© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.

  • Mus musculus (House mouse)
  • Cancer Research

Toxoplasma type II effector GRA15 has limited influence in vivo.

In PLoS ONE on 29 March 2024 by Merritt, E. F., Kochanowsky, J. A., et al.

Toxoplasma gondii is an intracellular parasite that establishes a long-term infection in the brain of many warm-blooded hosts, including humans and rodents. Like all obligate intracellular microbes, Toxoplasma uses many effector proteins to manipulate the host cell to ensure parasite survival. While some of these effector proteins are universal to all Toxoplasma strains, some are polymorphic between Toxoplasma strains. One such polymorphic effector is GRA15. The gra15 allele carried by type II strains activates host NF-κB signaling, leading to the release of cytokines such as IL-12, TNF, and IL-1β from immune cells infected with type II parasites. Prior work also suggested that GRA15 promotes early host control of parasites in vivo, but the effect of GRA15 on parasite persistence in the brain and the peripheral immune response has not been well defined. For this reason, we sought to address this gap by generating a new IIΔgra15 strain and comparing outcomes at 3 weeks post infection between WT and IIΔgra15 infected mice. We found that the brain parasite burden and the number of macrophages/microglia and T cells in the brain did not differ between WT and IIΔgra15 infected mice. In addition, while IIΔgra15 infected mice had a lower number and frequency of splenic M1-like macrophages and frequency of PD-1+ CTLA-4+ CD4+ T cells and NK cells compared to WT infected mice, the IFN-γ+ CD4 and CD8 T cell populations were equivalent. In summary, our results suggest that in vivo GRA15 may have a subtle effect on the peripheral immune response, but this effect is not strong enough to alter brain parasite burden or parenchymal immune cell number at 3 weeks post infection.
Copyright: © 2024 Merritt et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

  • Mus musculus (House mouse)
View this product on CiteAb