Product Citations: 108

Gut microbes play a crucial role in regulating the tumor microenvironment (TME) of colorectal cancer (CRC). Nevertheless, the deep mechanism between the microbiota-TME interaction has not been well explored. In this study, we for the first time discovered that Lactobacillus intestinalis (L. intestinalis) effectively suppressed tumor growth both in the AOM/DSS-induced CRC model and the ApcMin/+ spontaneous adenoma model. Our investigation revealed that L. intestinalis increased the infiltration of immune cells, particularly dendritic cells (DC), in the TME. Mechanically, the tumor-derived CCL5 induced by L. intestinalis recruited DC chemotaxis through the NOD1/NF-κB signaling pathway. In clinical samples and datasets, we found positive correlation between L. intestinalis, CCL5 level, and the DC-related genes. Our study provided a new strategy for microbial intervention for CRC and deepened the understanding of the interaction between tumor cells and the immune microenvironment modulated by gut microbes.

  • FC/FACS
  • Mus musculus (House mouse)
  • Cancer Research
  • Immunology and Microbiology

LAG3 limits regulatory T cell proliferation in α-synuclein gut-to-brain transmission model.

In Journal of Neuroinflammation on 5 July 2025 by Kong, W. X., Zhang, Z. L., et al.

Pathological α-synuclein (α-syn) can spread from the gut to the central nervous system (CNS), with CD4 + T cells playing a key role in this process. Lymphocyte activation gene 3 (LAG3) is involved in intestinal inflammation, regulates CD4 + T cell proliferation and function, and can specifically bind to pathological α-syn during cell-to-cell transmission. However, it remains unclear whether LAG3 is involved in the spread of pathological α-syn from the gut to the brain.
We utilized LAG3 knockout mice, combined with injection of α-syn preformed fibril (PFF) into the longitudinal and intermediate muscle layers of the pylorus and duodenum to model Parkinson's disease (PD). We used Immunohistochemistry staining, Western Blot, Flow cytometry to detect the changes of TH, α-syn, pro-inflammatory factors, barrier-related proteins and CD4 + T cells differentiation.
Our results show that LAG3 knockout partially alleviates psychological and behavioral deficits, dopamine system damage, and the gut-to-brain transmission of α-syn, which correlates with enhanced regulatory T cell (Treg) cell proliferation. Furthermore, LAG3 knockout improved intestinal dysfunction and increased the expression of tight junction proteins in both the gut and the blood-brain barrier (BBB). In CD4 + T cells isolated from the spleen, LAG3 knockout suppressed the aggregation of α-syn PFF, thereby inhibiting the toxic T-cell response induced by α-syn PFF. LAG3 deficiency also enhanced the IL-2/STAT5 signaling pathway, which regulates Treg proportions both in vivo and in vitro.
Our findings demonstrated that LAG3 intrinsically limits Treg cell proliferation and function in the environment with pathological α-syn and promotes the gut-to-brain transmission of α-syn.
© 2025. The Author(s).

  • Immunology and Microbiology

Activated immune cells infiltrate the vasculature during the pathophysiology of hypertension by establishing a vascular-immune interface that contributes to blood pressure dysregulation and organ failure. Many observations indicate a key role of CD8+ T cells in hypertension but mechanisms regulating their activation and interplay with the cardiovascular system are still unknown. In murine model, here we show that a specific member of the phosphoinositide-3-kinases (PI3K) family of lipid kinases, PI3Kγ, is a key intracellular signaling of CD8+ T cells activation and RANTES/CCL5 secretion in hypertension: CCL5-CCR5 signaling is crucial for the establishment of the vascular-immune interface in peripheral organs, lastly contributing to CD8+ tissue infiltration, organ dysfunction and blood pressure elevation. Our studies identify PI3Kγ as a booster of effector CD8+ T cell function, even in the absence of external stimuli. Lastly, an enhanced PI3Kγ signaling mediates the bystander activation of CD8+ T cells and proves effective in transferring the hypertensive phenotype between mice.
© 2025. The Author(s).

  • Cardiovascular biology
  • Immunology and Microbiology

Macrophage pyroptosis has been identified as a critical pathological mechanism in inflammation-related atherosclerosis (AS). In this work, we have demonstrated that Zn2+ features the strongest anti-inflammatory performance by screening 10 representative metal ions, and the MTC1 agonists can trigger lysosomal Zn2+ release and inhibit pyroptosis in macrophages. Based on these findings, we further engineered a mucolipin TRP channel 1 (MTC1)-related therapeutic nanoplatform for endogenously triggering lysosomal zinc release to curb inflammation and block macrophage pyroptosis. This nanoplatform consists of mesoporous silica nanoparticles to deliver MTC1 agonists and carbon nanodots, which could synergistically exert antiatherosclerotic effect by scavenging toxic reactive oxygen species, inhibiting macrophage pyroptosis, modulating macrophage transition, and rebuilding atherosclerotic immune microenvironment. These findings demonstrate that macrophage pyroptosis can be efficiently blocked via leveraging self-lysosomal zinc pool, which provides the paradigm of lysosomal zinc modulation-involved nanotherapeutics for managing other inflammatory diseases.

  • Cell Biology
  • Immunology and Microbiology

Following ischemic stroke, peripheral immune cell infiltration is characterized by myeloid cell predominance in the acute phase and lymphoid cell infiltration in the subacute to chronic phases. Endothelial cells, as a critical interface between the peripheral circulation and the brain, upregulate adhesion molecules to facilitate immune cell infiltration. However, it remains unclear whether endothelial cells exhibit functional differences at different stages after ischemic stroke and how these differences affect immune cell infiltration.
We performed single-cell RNA sequencing on peripheral immune and endothelial cells from Sham and middle cerebral artery occlusion (MCAO) mice at 3 and 14 days post-MCAO. Subsequent analysis of the sequencing data, combined with flow cytometry and immunofluorescence staining, was used to investigate the relationship between endothelial cell changes at different stages of stroke and immune cell infiltration.
We observed that the infiltration capacity of peripheral immune cells did not significantly increase at different stages after MCAO. However, endothelial cells underwent significant changes. By Day 3 post-MCAO, there was an increased proportion of venous endothelial cells with enhanced angiogenesis and adhesion functions. In this acute phase, newly formed venous endothelial cells with high expression of the adhesion molecule ICAM-1 were observed, promoting the infiltration of myeloid cells and NKT cells. From the acute to chronic phases, endothelial angiogenesis gradually decreased, accompanied by a marked increase in antigen presentation function. At 14 days post-MCAO, an increased proportion of VCAM-1-expressing venous endothelial cells was observed, potentially facilitating the infiltration of T cells and a subset of neutrophils. Furthermore, we discovered that the differential changes in venous endothelial cells at different stages after MCAO may be driven by distinct differentiation and proliferation patterns regulated by different signaling pathways.
Our study highlights that the differential expression of adhesion molecules and functional changes in endothelial cells at distinct stages after ischemic stroke may regulate the infiltration patterns of peripheral immune cells.
© 2025 The Author(s). CNS Neuroscience & Therapeutics published by John Wiley & Sons Ltd.

  • Cardiovascular biology
  • Immunology and Microbiology
View this product on CiteAb