peripheral immune cells
Recently Published Documents


TOTAL DOCUMENTS

170
(FIVE YEARS 121)

H-INDEX

20
(FIVE YEARS 6)

2022 ◽  
Vol 11 (2) ◽  
pp. 447
Author(s):  
Tomoko Takamatsu ◽  
Gaku Yamanaka ◽  
Koko Ohno ◽  
Kanako Hayashi ◽  
Yusuke Watanabe ◽  
...  

Neuroinflammation has been implicated in the pathogenesis of West syndrome (WS). Inflammatory cytokines, including interleukin-1β(IL-1β), have been reported to be associated with epilepsy. However, the assessment of cytokine changes in humans is not always simple or deterministic. This study aimed to elucidate the immunological mechanism of WS. We examined the intracellular cytokine profiles of peripheral blood cells collected from 13 patients with WS, using flow cytometry, and measured their serum cytokine levels. These were compared with those of 10 age-matched controls. We found that the WS group had significantly higher percentages of inter IL-1β, interleukin-1 receptor antagonist (IL-1RA)-positive monocytes, and interferon gamma (IFN-γ) in their CD8+ T cells than the control group. Interestingly, the group with sequelae revealed significantly lower levels of intracellular IFN-γ and IL-6 in their CD8+ T and CD4+ T cells, respectively, than the group without sequelae. There was no correlation between the ratios of positive cells and the serum levels of a particular cytokine in the WS patients. These cytokines in the peripheral immune cells might be involved in the neuroinflammation of WS, even in the absence of infectious or immune disease. Overall, an immunological approach using flow cytometry analysis might be useful for immunological studies of epilepsy.


2022 ◽  
Vol 5 (1) ◽  
Author(s):  
Madalena Cipriano ◽  
Katharina Schlünder ◽  
Christopher Probst ◽  
Kirstin Linke ◽  
Martin Weiss ◽  
...  

AbstractDisorders of the eye leading to visual impairment are a major issue that affects millions of people. On the other side ocular toxicities were described for e.g. molecularly targeted therapies in oncology and may hamper their development. Current ocular model systems feature a number of limitations affecting human-relevance and availability. To find new options for pharmacological treatment and assess mechanisms of toxicity, hence, novel complex model systems that are human-relevant and readily available are urgently required. Here, we report the development of a human immunocompetent Choroid-on-Chip (CoC), a human cell-based in vitro model of the choroid layer of the eye integrating melanocytes and microvascular endothelial cells, covered by a layer of retinal pigmented epithelial cells. Immunocompetence is achieved by perfusion of peripheral immune cells. We demonstrate controlled immune cell recruitment into the stromal compartments through a vascular monolayer and in vivo-like cytokine release profiles. To investigate applicability for both efficacy testing of immunosuppressive compounds as well as safety profiling of immunoactivating antibodies, we exposed the CoCs to cyclosporine and tested CD3 bispecific antibodies.


2022 ◽  
Vol 12 ◽  
Author(s):  
Jing Zhang ◽  
Zi Li ◽  
Huijun Lu ◽  
Junchao Shi ◽  
Rui Gao ◽  
...  

Porcine hemagglutinating encephalomyelitis virus (PHEV) is a highly neurotropic coronavirus that invades the host central nervous system (CNS) and causes neurological dysfunction. Microglia are key immune cells in the CNS, however, whether and how they response to PHEV infection remains unclear. Herein, microglial activation and proliferation were detected in the CNS of PHEV-infected mice, as along with the proinflammatory response. Moreover, the production of proinflammatory cytokines induced by moderately activated microglia limited viral replication in the early stage of infection. Microglial depletion assays showed that during late infection, excess activation of microglia aggravated neurological symptoms, BBB destruction, and peripheral monocyte/macrophage infiltration into the CNS. Using an in vitro brain slice model, PHEV was identified to specifically and moderately induce microglial activation in the absence of peripheral immune cells infiltration. Consistently, macrophage clearance from circulating blood indicated that peripheral monocytes/macrophages crossing the BBB of mice were responsible for excess activation of microglia and CNS damage in late PHEV infection. Overall, our findings provide evidence supporting a dual role for microglia in the host CNS in response to coronavirus PHEV invasion.


2022 ◽  
Vol 15 ◽  
Author(s):  
Nynke J. van den Hoogen ◽  
Erika K. Harding ◽  
Chloé E. D. Davidson ◽  
Tuan Trang

Chronic pain is a complex sensory, cognitive, and emotional experience that imposes a great personal, psychological, and socioeconomic burden on patients. An estimated 1.5 billion people worldwide are afflicted with chronic pain, which is often difficult to treat and may be resistant to the potent pain-relieving effects of opioid analgesics. Attention has therefore focused on advancing new pain therapies directed at the cannabinoid system because of its key role in pain modulation. Endocannabinoids and exogenous cannabinoids exert their actions primarily through Gi/o-protein coupled cannabinoid CB1 and CB2 receptors expressed throughout the nervous system. CB1 receptors are found at key nodes along the pain pathway and their activity gates both the sensory and affective components of pain. CB2 receptors are typically expressed at low levels on microglia, astrocytes, and peripheral immune cells. In chronic pain states, there is a marked increase in CB2 expression which modulates the activity of these central and peripheral immune cells with important consequences for the surrounding pain circuitry. Growing evidence indicate that interventions targeting CB1 or CB2 receptors improve pain outcomes in a variety of preclinical pain models. In this mini-review, we will highlight recent advances in understanding how cannabinoids modulate microglia function and its implications for cannabinoid-mediated analgesia, focusing on microglia-neuron interactions within the spinal nociceptive circuitry.


2021 ◽  
Author(s):  
Lexing Xie ◽  
Shuang Zhang ◽  
Li Huang ◽  
Zhouzhou Peng ◽  
Hui Lu ◽  
...  

Abstract Background: Stroke persists as a major cause of morbidity and mortality worldwide. After a stroke, peripheral immune cells are rapidly activated and then infiltrate the central nervous system to cause inflammation in the brain. However, it is not clear when and how these peripheral immune cells affect the central inflammatory response and whether there are intervention targets that can alleviate ischemia-reperfusion injury. In this study, we collected mouse peripheral blood samples at different time points after stroke for single-cell sequencing to reveal the dynamic changes in peripheral immune cells. Methods: We performed single-cell sequencing on peripheral blood of mice at 1, 3, 7, and 14 days after ischemia-reperfusion to analyze the changes of subpopulations after cerebral ischemia-reperfusion; Real-time PCR, western blot and enzyme‑linked immunoabsorbent assay were used to perform mRNA and protein levels verification; Immunoprecipitation verifies the interaction of proteins and between junctional adhesion molecule (JAM-A) and Cathepsin S (CTSS) protein, in vitro enzyme digestion and silver staining method to detect the protease digestion effect of CTSS.Results: Peripheral monocyte subpopulations increased significantly after ischemia-reperfusion. Pseudo-time trajectory analysis and gene function analysis further suggested that CTSS may play an important role in regulating monocyte activation and leading to proteolysis. Next, we found that the expression of CTSS was significantly increased in monocytes after I/R in mice. Then, we used CTSS inhibitors and knockout mouse experiments to prove that inhibiting its expression can significantly reduce infarct volume and reduce blood–brain barrier (BBB) leakage. In addition, we found that CTSS destroys BBB by binding to JAM family proteins to cause them degradation. Conclusion: Inhibition of Cathepsin S attenuated cerebral ischemia reperfusion injury and Cathepsin S can be used as a novel target for drug intervention after stroke.


2021 ◽  
Vol 15 ◽  
Author(s):  
Carlos del Pilar ◽  
Rafael Lebrón-Galán ◽  
Ester Pérez-Martín ◽  
Laura Pérez-Revuelta ◽  
Carmelo Antonio Ávila-Zarza ◽  
...  

The progression of neurodegenerative diseases is reciprocally associated with impairments in peripheral immune responses. We investigated different contexts of selective neurodegeneration to identify specific alterations of peripheral immune cells and, at the same time, discover potential biomarkers associated to this pathological condition. Consequently, a model of human cerebellar degeneration and ataxia -the Purkinje Cell Degeneration (PCD) mouse- has been employed, as it allows the study of different processes of selective neuronal death in the same animal, i.e., Purkinje cells in the cerebellum and mitral cells in the olfactory bulb. Infiltrated leukocytes were studied in both brain areas and compared with those from other standardized neuroinflammatory models obtained by administering either gamma radiation or lipopolysaccharide. Moreover, both myeloid and lymphoid splenic populations were analyzed by flow cytometry, focusing on markers of functional maturity and antigen presentation. The severity and type of neural damage and inflammation affected immune cell infiltration. Leukocytes were more numerous in the cerebellum of PCD mice, being located predominantly within those cerebellar layers mostly affected by neurodegeneration, in a completely different manner than the typical models of induced neuroinflammation. Furthermore, the milder degeneration of the olfactory bulb did not foster leukocyte attraction. Concerning the splenic analysis, in PCD mice we found: (1) a decreased percentage of several myeloid cell subsets, and (2) a reduced mean fluorescence intensity in those myeloid markers related to both antigen presentation and functional maturity. In conclusion, the selective degeneration of Purkinje cells triggers a specific effect on peripheral immune cells, fostering both attraction and functional changes. This fact endorses the employment of peripheral immune cell populations as concrete biomarkers for monitoring different neuronal death processes.


Antioxidants ◽  
2021 ◽  
Vol 10 (12) ◽  
pp. 1917
Author(s):  
Yuanxin Zhao ◽  
Buhan Liu ◽  
Long Xu ◽  
Sihang Yu ◽  
Jiaying Fu ◽  
...  

One of the most striking hallmarks shared by various neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease and amyotrophic lateral sclerosis, is microglia-mediated and astrocyte-mediated neuroinflammation. Although inhibitions of both harmful proteins and aggregation are major treatments for neurodegenerative diseases, whether the phenomenon of non-normal protein or peptide aggregation is causally related to neuronal loss and synaptic damage is still controversial. Currently, excessive production of reactive oxygen species (ROS), which induces mitochondrial dysfunction in neurons that may play a key role in the regulation of immune cells, is proposed as a regulator in neurological disorders. In this review, we propose that mitochondrial DNA (mtDNA) release due to ROS may act on microglia and astrocytes adjacent to neurons to induce inflammation through activation of innate immune responses (such as cGAS/STING). Elucidating the relationship between mtDNA and the formation of a pro-inflammatory microenvironment could contribute to a better understanding of the mechanism of crosstalk between neuronal and peripheral immune cells and lead to the development of novel therapeutic approaches to neurodegenerative diseases.


Author(s):  
Yaping Liu

AbstractEpigenetic mechanisms play instrumental roles in gene regulation during embryonic development and disease progression. However, it is challenging to non-invasively monitor the dynamics of epigenomes and related gene regulation at inaccessible human tissues, such as tumours, fetuses and transplanted organs. Circulating cell-free DNA (cfDNA) in peripheral blood provides a promising opportunity to non-invasively monitor the genomes from these inaccessible tissues. The fragmentation patterns of plasma cfDNA are unevenly distributed in the genome and reflect the in vivo gene-regulation status across multiple molecular layers, such as nucleosome positioning and gene expression. In this review, we revisited the computational and experimental approaches that have been recently developed to measure the cfDNA fragmentomics across different resolutions comprehensively. Moreover, cfDNA in peripheral blood is released following cell death, after apoptosis or necrosis, mainly from haematopoietic cells in healthy people and diseased tissues in patients. Several cfDNA-fragmentomics approaches showed the potential to identify the tissues-of-origin in cfDNA from cancer patients and healthy individuals. Overall, these studies paved the road for cfDNA fragmentomics to non-invasively monitor the in vivo gene-regulatory dynamics in both peripheral immune cells and diseased tissues.


2021 ◽  
Vol 12 ◽  
Author(s):  
Yue Zhou ◽  
Xuelian Liao ◽  
Xiangrong Song ◽  
Min He ◽  
Fei Xiao ◽  
...  

During the COVID-19 pandemic, a phenomenon emerged in which some patients with severe disease were critically ill and could not be discharged from the ICU even though they exhibited negative viral tests. To explore the underlying mechanism, we collected blood samples from these patients and analyzed the gene expression profiles of peripheral immune cells. We found that all enrolled patients, regardless of changes in genes related to different symptoms and inflammatory responses, showed universally and severely decreased expression of adaptive immunity-related genes, especially those related to T/B cell arms and HLA molecules, and that these patients exhibited long-term secondary infections. In addition, no significant change was found in the expression of classic immunosuppression molecules including PD-1, PD-L1, and CTLA-4, suggesting that the adaptive immune suppression may not be due to the change of these genes. According to the published literatures and our data, this adaptive immunosuppression is likely to be caused by the “dysregulated host response” to severe infection, similar to the immunosuppression that exists in other severely infected patients with sepsis.


Sign in / Sign up

Export Citation Format

Share Document