Product Citations: 20

Acute neuronal cell death and neuroinflammation per se do not trigger secondary autoimmune encephalitis in mice.

In Scientific Reports on 27 June 2025 by Wilke, J., Ntolkeras, A., et al.

Patients with virus encephalitis, such as herpes simplex encephalitis and Japanese encephalitis frequently relapse with autoimmune encephalitides associated with neural autoantibodies. It has been hypothesized that the infection-induced damage to the central nervous system results in shedding of neural autoantigens, their presentation to the peripheral immune system, and initiation of a secondary autoimmune encephalitis that targets these autoantigens. To test this hypothesis, we utilized a transgenic mouse model of virus-like but sterile encephalitis. After induction of acute neuronal death in the hippocampus, we monitored the mice for encephalitis-like symptoms for up to 10 months, evaluated the degree of neuroinflammation at several time points and screened their plasma for autoantibodies against 49 different autoimmune disease-associated brain autoantibodies. Throughout the study period, we did not detect any symptoms of severe autoimmune encephalitis, like hyperactivity, circling, seizures, lethargy. Evaluation of microglia numbers and morphology revealed pronounced microgliosis 1-week after initial encephalitis induction, which decreased over time. Scattered lymphocyte infiltration was present at all times in hippocampi of encephalitis mice, and did not increase over time. Perivascular cuffs were not detected. Infiltrating lymphocytes mainly consisted of CD8+ T cells. B cell infiltration was rare and did not differ from healthy control mice. High-parameter immunophenotyping of peripheral blood leukocytes did not reveal any changes associated with an autoimmune response. Testing all plasma samples (n = 30/group) at a dilution of 1:100 for autoantibodies against 49 neural autoantigens gave only two positive results, namely one healthy control with anti-CASPR2 autoantibodies (IgG) and one post-encephalitis mouse with anti-homer-3 autoantibodies (IgM). Overall, these findings suggest that acute neuronal cell death and neuroinflammation per se are not sufficient to trigger downstream autoimmune encephalitis relapses in mice.
© 2025. The Author(s).

  • IHC
  • Immunology and Microbiology

The lack of T cells in tumors is a major hurdle to successful immune checkpoint therapy (ICT). Therefore, therapeutic strategies promoting T cell recruitment into tumors are warranted to improve the treatment efficacy. Here, we report that Fc-optimized anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies are potent remodelers of tumor vasculature that increase tumor-associated high endothelial venules (TA-HEVs), specialized blood vessels supporting lymphocyte entry into tumors. Mechanistically, this effect is dependent on the Fc domain of anti-CTLA-4 antibodies and CD4+ T cells and involves interferon gamma (IFNγ). Unexpectedly, we find that the human anti-CTLA-4 antibody ipilimumab fails to increase TA-HEVs in a humanized mouse model. However, increasing its Fc effector function rescues the modulation of TA-HEVs, promotes CD4+ and CD8+ T cell infiltration into tumors, and sensitizes recalcitrant tumors to programmed cell death protein 1 (PD-1) blockade. Our findings suggest that Fc-optimized anti-CTLA-4 antibodies could be used to reprogram tumor vasculature in poorly immunogenic cold tumors and improve the efficacy of ICT.
Copyright © 2025 The Author(s). Published by Elsevier Inc. All rights reserved.

  • Cancer Research

Study of Effector CD8+ T Cell Interactions with Cortical Neurons in Response to Inflammation in Mouse Brain Slices and Neuronal Cultures.

In International Journal of Molecular Sciences on 6 February 2023 by Lin, C. H., Scheller, A., et al.

Cytotoxic CD8+ T cells contribute to neuronal damage in inflammatory and degenerative CNS disorders, such as multiple sclerosis (MS). The mechanism of cortical damage associated with CD8+ T cells is not well understood. We developed in vitro cell culture and ex vivo brain slice co-culture models of brain inflammation to study CD8+ T cell-neuron interactions. To induce inflammation, we applied T cell conditioned media, which contains a variety of cytokines, during CD8+ T cell polyclonal activation. Release of IFNγ and TNFα from co-cultures was verified by ELISA, confirming an inflammatory response. We also visualized the physical interactions between CD8+ T cells and cortical neurons using live-cell confocal imaging. The imaging revealed that T cells reduced their migration velocity and changed their migratory patterns under inflammatory conditions. CD8+ T cells increased their dwell time at neuronal soma and dendrites in response to added cytokines. These changes were seen in both the in vitro and ex vivo models. The results confirm that these in vitro and ex vivo models provide promising platforms for the study of the molecular details of neuron-immune cell interactions under inflammatory conditions, which allow high-resolution live microscopy and are readily amenable to experimental manipulation.

  • Immunology and Microbiology
  • Neuroscience

Senescence Connects Autophagy Deficiency to Inflammation and Tumor Progression in the Liver.

In Cellular and Molecular Gastroenterology and Hepatology on 11 April 2022 by Huda, N., Khambu, B., et al.

Cellular senescence frequently is present in injured livers. The induction mechanism and the pathologic role are not always clear. We aimed to understand the dynamics of senescence induction and progression, and the mechanism responsible for the pathology using a mouse model that disables the essential process of autophagy.
Mice deficient in key autophagy genes Atg7 or Atg5 in the liver were used. Senescence was measured using established cellular and molecular signatures. The mechanistic roles of nuclear factor erythroid 2 (NRF2), forkhead box K1, and C-C motif chemokine receptor 2 (CCR2) were assessed using mouse genetic models. Liver functions, pathology, and tumor development were measured using biochemical and histologic approaches.
Inducible deletion of Atg7 rapidly up-regulated cyclin-dependent kinase inhibitors independently of injury and induced senescence-associated β-galactosidase activities and senescence-associated secretory phenotype (SASP). Sustained activation of NRF2 was the major factor causing senescence by mediating oxidative DNA damage and up-regulating C-C motif chemokine ligand 2, a key component of autophagy-related SASP, via the NRF2-forkhead box K1 axis. Senescence was responsible for hepatic inflammation through CCR2-mediated recruitment of CD11b+ monocytes and CD3+ T cells. The CCR2-mediated process in turn enhanced senescence and SASP by up-regulating cyclin-dependent kinase inhibitors and chemokines. Thus, senescence and inflammation can mutually augment each other, forming an amplification loop for both events. The CCR2-mediated process also modulated liver injury and tumor progression at the later stage of autophagy deficiency-related pathology.
These results provide the insight that hepatic senescence can occur early in the disease process, triggers inflammation and is enhanced by inflammation, and has long-term effects on liver injury and tumor progression.
Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

  • IHC
  • Mus musculus (House mouse)
  • Cancer Research
  • Cell Biology
  • Immunology and Microbiology

To improve the potential treatment strategies of incurable renal cell carcinoma (RCC), which is highly resistant to chemotherapy and radiotherapy, the present study established a combination therapy with immunostimulatory factor (ISTF) and anti-4-1BB monoclonal antibodies (mAbs) to augment the antitumor response in a murine RCC model. ISTF isolated from Actinobacillus actinomycetemcomitans stimulates macrophages, dendritic cells and B cells to produce IL-6, TNF-α, nitric oxide and major histocompatibility complex class II expression. 4-1BB (CD137) is expressed in activated immune cells, including activated T cells, and is a promising target for cancer immunotherapy. The administration of anti-4-1BB mAbs promoted antitumor immunity via enhancing CD11c+CD8+ T cells. The CD11c+CD8+ T cells were characterized by high killing activity and IFN-γ-producing ability, representing a phenotype of active effector cytotoxic T lymphocytes. The present study showed that combination therapy with ISTF and anti-4-1BB mAbs promoted partial tumor regression with established RCC, but monotherapy with ISTF or anti-4-1BB mAbs did not. These effects were speculated to be caused by the increase in CD11c+CD8+ T cells in the spleen and tumor, and IFN-γ production. These insights into the effector mechanisms of the combination of ISTF and anti-4-1BB mAbs may be useful for targeting incurable RCC.
Copyright: © Ju et al.

  • Cancer Research
  • Immunology and Microbiology
View this product on CiteAb