Product Citations: 44

FLT1 activation in cancer cells promotes PARP-inhibitor resistance in breast cancer.

In EMBO Molecular Medicine on 1 August 2024 by Tai, Y., Chow, A., et al.

Acquired resistance to PARP inhibitors (PARPi) remains a treatment challenge for BRCA1/2-mutant breast cancer that drastically shortens patient survival. Although several resistance mechanisms have been identified, none have been successfully targeted in the clinic. Using new PARPi-resistance models of Brca1- and Bard1-mutant breast cancer generated in-vivo, we identified FLT1 (VEGFR1) as a driver of resistance. Unlike the known role of VEGF signaling in angiogenesis, we demonstrate a novel, non-canonical role for FLT1 signaling that protects cancer cells from PARPi in-vivo through a combination of cell-intrinsic and cell-extrinsic pathways. We demonstrate that FLT1 blockade suppresses AKT activation, increases tumor infiltration of CD8+ T cells, and causes dramatic regression of PARPi-resistant breast tumors in a T-cell-dependent manner. Moreover, PARPi-resistant tumor cells can be readily re-sensitized to PARPi by targeting Flt1 either genetically (Flt1-suppression) or pharmacologically (axitinib). Importantly, a retrospective series of breast cancer patients treated with PARPi demonstrated shorter progression-free survival in cases with FLT1 activation at pre-treatment. Our study therefore identifies FLT1 as a potential therapeutic target in PARPi-resistant, BRCA1/2-mutant breast cancer.
© 2024. The Author(s).

  • Biochemistry and Molecular biology
  • Cancer Research

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

Myeloid dickkopf-1 fuels neurovascular and neuroimmune alterations in ischemic stroke

Preprint on Research Square on 14 August 2023 by ElAli, A., Menet, R., et al.

Neurovascular impairments and neuroimmune deregulation contribute to injury progression after ischemic stroke. Dickkopf-1 (DKK1) elevated levels correlates with poor stroke outcomes. DKK1 antagonizes the canonical Wnt pathway that plays a critical role in regulating neurovascular and neuroimmune functions. Herein, we report that DKK1 expression in the normal adult brain is absent, but is de novo expressed at the lesion site after experimental ischemic stroke. Using genetic tools to conditionally induce DKK1 expression in a tissue-specific manner, we reveal that its early induction aggravates neurological deficits and injury severity after stroke, associated with altered neuronal and vascular functions. DKK1 post-stroke induction hinders lesion containment by disorganizing the astroglial scar, leading to a chronic neuroinflammation and increased anxiety-like behaviors. Using chimeric mice, we unravel thatDKK1 is released by bone marrow-derived cells (BMDCs) expressing myeloid markers that infiltrate the lesion site. DKK1 restricted induction in BMDCs is sufficient to mediate astroglial scar disorganization. Notably, neutralization of DKK1 limits injury progression and improves neurological recovery after stroke. Our findings indicate that BMDCs-derived DKK1 promotes injury progression after stroke and suggest that neutralizing its biological activity represent a promising therapeutic avenue for ischemic stroke.

  • Mus musculus (House mouse)
  • Cardiovascular biology

Theiler's murine encephalomyelitis virus (TMEV) induces an acute polioencephalomyelitis and a chronic demyelinating leukomyelitis in SJL mice. C57BL/6 (B6) mice generally do not develop TMEV-induced demyelinating disease (TMEV-IDD) due to virus elimination. However, TMEV can persist in specific immunodeficient B6 mice such as IFNβ-/- mice and induce a demyelinating process. The proinflammatory cytokines IL-1β and IL-18 are activated by the inflammasome pathway, which consists of a pattern recognition receptor molecule sensing microbial pathogens, the adaptor molecule Apoptosis-associated speck-like protein containing a CARD (ASC), and the executioner caspase-1. To analyze the contribution of the inflammasome pathway to the resistance of B6 mice to TMEV-IDD, ASC- and caspase-1-deficient mice and wild type littermates were infected with TMEV and investigated using histology, immunohistochemistry, RT-qPCR, and Western Blot. Despite the antiviral activity of the inflammasome pathway, ASC- and caspase-1-deficient mice eliminated the virus and did not develop TMEV-IDD. Moreover, a similar IFNβ and cytokine gene expression was found in the brain of immunodeficient mice and their wild type littermates. Most importantly, Western Blot showed cleavage of IL-1β and IL-18 in all investigated mice. Consequently, inflammasome-dependent activation of IL-1β and IL-18 does not play a major role in the resistance of B6 mice to TMEV-IDD.
© 2023. The Author(s).

  • Mus musculus (House mouse)
  • Immunology and Microbiology

Stress promotes the infiltration of peripheral immune cells to the brain.

In Brain, Behavior, and Immunity on 1 July 2023 by Medina-Rodríguez, E. M., Han, D., et al.

Immune cells and the brain have a privileged interaction. Here, we report changes in the hippocampal immune microenvironment at the single cell level after stress, uncovering the tight orchestration of immune cell infiltration into the hippocampus after stress to maintain homeostasis. We show the distribution of several immune cell types in the hippocampus associated with their susceptibility or resilience to the learned helplessness paradigm in a sex- and microbiota-dependent manner using single-cell RNA sequencing and bioinformatic tools, flow cytometry, and immunofluorescence. We uncovered the presence of tissue-resident memory T cells that accumulate over time in the hippocampus of learned helpless mice, and the presence of CD74-expressing myeloid cells. These cells were found by a knockdown approach to be critical to induce resilience to learned helplessness. Altogether, these findings provide a novel overview of the neuro-immune repertoire and its impact on the landscape of the hippocampus after learned helplessness.
Copyright © 2023 Elsevier Inc. All rights reserved.

  • Immunology and Microbiology
View this product on CiteAb