scholarly journals IL-4 induces a wide-spectrum intracellular signaling cascade in CD8+T cells

2007 ◽  
Vol 81 (4) ◽  
pp. 1102-1110 ◽  
Author(s):  
Ana Acacia de Sa Pinheiro ◽  
Alexandre Morrot ◽  
Sumana Chakravarty ◽  
Michael Overstreet ◽  
Jay H. Bream ◽  
...  
FACE ◽  
2021 ◽  
pp. 273250162110243
Author(s):  
Mikhail Pakvasa ◽  
Andrew B. Tucker ◽  
Timothy Shen ◽  
Tong-Chuan He ◽  
Russell R. Reid

Hedgehog signaling was discovered more than 40 years ago in experiments demonstrating that it is a fundamental mediator of limb development. Since that time, it has been shown to be important in development, homeostasis, and disease. The hedgehog pathway proceeds through a pathway highly conserved throughout animals beginning with the extracellular diffusion of hedgehog ligands, proceeding through an intracellular signaling cascade, and ending with the activation of specific target genes. A vast amount of research has been done elucidating hedgehog signaling mechanisms and regulation. This research has found a complex system of genetics and signaling that helps determine how organisms develop and function. This review provides an overview of what is known about hedgehog genetics and signaling, followed by an in-depth discussion of the role of hedgehog signaling in craniofacial development and carcinogenesis.


2009 ◽  
Vol 207 (1) ◽  
pp. 207-221 ◽  
Author(s):  
Cláudia C. Oliveira ◽  
Peter A. van Veelen ◽  
Bianca Querido ◽  
Arnoud de Ru ◽  
Marjolein Sluijter ◽  
...  

The nonclassical major histocompatibility complex (MHC) Qa-1b accommodates monomorphic leader peptides and functions as a ligand for germ line receptors CD94/NKG2, which are expressed by natural killer cells and CD8+ T cells. We here describe that the conserved peptides are replaced by a novel peptide repertoire of surprising diversity as a result of impairments in the antigen-processing pathway. This novel peptide repertoire represents immunogenic neoantigens for CD8+ T cells, as we found that these Qa-1b–restricted T cells dominantly participated in the response to tumors with processing deficiencies. A surprisingly wide spectrum of target cells, irrespective of transformation status, MHC background, or type of processing deficiency, was recognized by this T cell subset, complying with the conserved nature of Qa-1b. Target cell recognition depended on T cell receptor and Qa-1b interaction, and immunization with identified peptide epitopes demonstrated in vivo priming of CD8+ T cells. Our data reveal that Qa-1b, and most likely its human homologue human leukocyte antigen-E, is important for the defense against processing-deficient cells by displacing the monomorphic leader peptides, which relieves the inhibition through CD94/NKG2A on lymphocytes, and by presenting a novel repertoire of immunogenic peptides, which recruits a subset of cytotoxic CD8+ T cells.


2009 ◽  
Vol 44 (4) ◽  
pp. 462-470 ◽  
Author(s):  
Cun-Shuan Xu ◽  
Heng-Yi Shao ◽  
Shuai-Shuai Liu ◽  
Bo Qin ◽  
Xiu-Feng Sun ◽  
...  

2021 ◽  
Author(s):  
◽  
Wei Su ◽  

Inflammation is essential for the clearance of pathogens and to facilitate healing of damaged tissue. However, this process must be controlled to limit immunopathology. Cell-intrinsic effects of inhibitory and signaling molecules are known to maintain quiescence and prevent effector differentiation and inflammation. Moreover, specific populations of immune cells exert cell-extrinsic effects for immunosuppression. Therefore, studies on the immunosuppressive functions of these cell populations will provide a better understanding of how inflammation is regulated and how its dysregulation causes human disease. Additional insights in this area may uncover novel targets to be manipulated for therapeutic benefit in autoimmune and inflammatory disorders, such as neurodegenerative diseases. Foxp3-expressing regulatory T (Treg) cells are specialized immunosuppressive cells that establish immune tolerance to prevent the development of autoimmune and other inflammatory diseases, with effector-Treg (eTreg) cells playing a pivotal role. Recently, cellular metabolism has emerged as a mediator to enforce Treg-cell function and heterogeneity. In Chapter 3, we used genetic and pharmacological tools to demonstrate that isoprenoid-dependent posttranslational lipid modifications dictate eTreg-cell accumulation and function by intersecting with T cell receptor (TCR)-induced intracellular signaling. We showed that isoprenoids are essential for activated Treg-cell suppressive activity, and Treg cell-specific deletion of the enzymes that mediate farnesylation and geranylgeranylation (encoded by Fntb and Pggt1b, respectively) leads to a reduction of eTreg cells and the development of fatal autoimmunity. In Chapter 4, we further explored the mechanistic role of protein prenylation in the regulation of eTreg cells by performing a comprehensive analysis of protein prenylation-dependent molecular signaling in eTreg-cells. Specifically, we found that Fntb drives eTreg-cell maintenance by promoting mTORC1 activity-dependent proliferation and ICOS-mediated cellular fitness. In contrast, Pggt1b orchestrated transcriptional programming by TCR stimulation and Rac signaling to establish eTreg-cell differentiation and immune tolerance. Therefore, our results reveal a bidirectional interplay between immune signals, metabolism-mediated posttranslational modifications, and intracellular signaling for the differentiation and maintenance of eTreg cells. Neuroinflammation is also a feature of neurogenerative diseases, but the underlying cellular mechanisms that limit inflammation in neurodegenerative diseases are largely unknown. In Chapter 5, using single cell RNA-sequencing (scRNA-seq) of immune cells in a mouse model of neurodegeneration (specifically, Alzheimer’s disease AD), we found that CD8 T cells accumulated in the brain parenchyma. These CD8 T cells had tissue resident memory-associated features and appeared to retain functionality. Importantly, T cell ablation was found to exacerbate disease-associated deposition of Beta- amyloid (A-Beta) and cognitive decline in a mouse model of AD. Moreover, in the absence of T cells, microglia acquired proinflammatory features and clustered in regions near A-Beta plaques, features associated with more severe disease. Collectively, these results suggest that T cells are critical to restrain microglia activation and limit neurodegeneration-associated pathologies in a murine model of AD.


Hypertension ◽  
2016 ◽  
Vol 68 (suppl_1) ◽  
Author(s):  
Daniela Carnevale ◽  
Maria Piacenti ◽  
Giuseppe Cifelli ◽  
Roberta Iacobucci ◽  
Giuseppe Lembo

In the field of research exploring the connection existing between hypertension and immune system, CD8 effector T cells emerge as the possible mediators of target organ colonization. In the absence of overt inflammation or pathogen response, naïve T cells circulate from the blood into secondary lymphoid organs, where, upon challenge, become activated. Then, they differentiate into effector T cells, which display typical activation patterns. However, less is known about the intracellular signaling pathways that are responsible for the acquisition of effectors functions in T cells. The p110γ isoform of the PI3K family has unique features, being crucially involved in the immune and cardiovascular systems. On this issue, we have described that PI3Kγ has a crucial role in blood pressure regulation, being KO mice protected from AngII-induced hypertension. Moreover, we found that mice with a constitutively active PI3Kγ isoform (CAAX mice) were spontaneously hypertensive (SBP: CAAX 135 ± 3 vs WT 105 ± 4 mmHg, p<0.001). Interestingly, PI3Kγ is known to play a selective role in regulating the migration of effector CD8 T cells, even though there was no effect of PI3Kγ in naïve T cells. Thus we explored the possible involvement of PI3Kγ in the crosstalk between hypertension and immunity. CAAX mice displayed a significant infiltration of activated CD8 + CD69 + T cells in kidney, as compared to WT mice (10.2 ± 2.1 vs 2.8 ± 0.6 *10 4 cells/kidney, p<0.01). At the functional level, this phenotype was associated with enlarged Bowman’s spaces and fibrosis in the kidney of CAAX mice, leading to disruption of renal function, as shown by later development of proteinuria. In the end, to demonstrate whether the CAAX hypertensive phenotype, associated to renal damage after CD8 colonization, could be ascribed to the overactivation of PI3Kγ signaling in this immune cell type, we performed an adoptive transfer of CD8 T cells isolated from CAAX mice in WT mice. Strikingly we found that CD8 T cells with constitutively active PI3Kγ were effective to induce hypertension in naïve mice. These data suggest that in the development of hypertension, PI3Kγ signaling in CD8 T cells is crucial for their accumulation in the kidney, likely contributing to increase in blood pressure by altering renal function.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2680-2680
Author(s):  
Kathryn I Sunthankar ◽  
Benjamin J Reisman ◽  
Candace H Cote ◽  
Paul Brent Ferrell

In healthy hematopoiesis, cell identity and signaling response are tightly linked, with predictable cell type-specific responses to cytokines and growth factors. However, this correlation is often disrupted in myeloid malignancies, including acute myeloid leukemia (AML), wherein signaling responses may be driven directly by kinase mutational activation or cell state changes due to epigenetic alterations, for instance. In order to resolve ligand-driven signaling pathways in bone marrow, tools that allow simultaneous phenotypic characterization and functional cellular responses at single cell resolution are needed. Here, we present the use of a high content mass cytometry panel combined with mass-tag cell barcoding in order to characterize cell identity and signaling responses in bone marrow hematopoietic cells from healthy donors and leukemic patients. We created a phospho-specific mass cytometry panel comprising 24 surface phenotyping markers to resolve the predominant cellular subsets within bone marrow and blood. We perturbed cellular signaling with nine growth factors, cytokines, and chemicals and measured immediate (15 minute) responses at 10 intracellular signaling markers (pSTAT1, pSTAT3, pSTAT5, pSYK, pp38, pERK1/2, pS6, pNFkB, IkBa, & pAKT). To improve robustness of the signaling response analysis, we used mass-tag cell barcoding with palladium prior to surface and intracellular antibody staining, followed by computational debarcoding. Downstream analysis was performed with Cytobank and R. The data set included thirty-five AML patient samples and seven healthy controls with greater than 300,000 cells collected over the 10 barcoded conditions (unstimulated and 9 stimulation conditions). Dimensionality reduction with uniform manifold approximation and projection (UMAP) combined with topological clustering (HDBSCAN) enabled initial data analysis and was followed by expert identification of resultant clusters via surface marker expression. Density-based clustering of the common UMAP embedding of all samples identified known subsets of hematopoietic cells (B cells, CD4 (CD25+ and CD25-) and CD8 T cells, double negative (DN) T cells, NK cells (three subsets), erythroblasts, several subsets of myeloid and leukemia cells, and hematopoietic stem cells (HSCs)). Mass-tag cell barcoding provided stable UMAP embeddings for each sample over the 10 stimulation conditions. High dimensional signaling response was calculated per cell and per each major cell subset for the 90 nodes (9 conditions by 10 markers) and hierarchical clustering stratified samples based upon signaling signatures. Signaling responses varied across non-leukemia and leukemia cell populations in AML samples, whereas cellular phenotypes were more well correlated with signaling phenotypes in healthy samples. Heterogeneity in signaling response was driven by variability seen in several "stimulation:response" pairs. The most impactful pairs to clustering of AML blasts were IFNγ:pSTAT1, GM-CSF:pSTAT5, IL-3:pSTAT5, PMA:pS6, and IL-6:pSTAT3. Favorable risk samples (by European LeukemiaNet risk stratification) were found to have significantly larger pSTAT5 increases to IL-3 and GM-CSF than both intermediate and adverse risk subgroups. In CD8 T cells, responsiveness to PMA and IL-10 drove clustering, and, in particular, samples with ELN adverse risk showed reduced PMA:pS6 and PMA:pERK responses. We present a robust evaluation of intracellular signaling responses in the bone marrow cellular environment of AML. These data provide rationale for ongoing investigation aimed at targeting both leukemia and non-leukemia cell signaling pathways in the treatment of AML. Disclosures Ferrell: Incyte: Research Funding; Forma Therapeutics: Research Funding; Agios: Consultancy; Astex Pharmaceuticals: Research Funding.


Author(s):  
Tzyy-Yue Wong ◽  
Yu-Kai Tseng ◽  
Tung-Chen Yeh ◽  
Rong-Chang Jhong ◽  
Yue-Fang Wang ◽  
...  

Thought runs through the mind like blood runs through our body to keep us alive. Like the mind, the body does not stay inert and is in constant motion. Not a single cell in our body is left inert unless cell is under stress or dying. These scenarios are reflected upon when a person is sick, the person lies in bed with less movement; however, is active when the person is healthy. The topic of mechanical stimulation has emerged due to the increasing understanding of the physical stimulations we face each day. Further understanding of the mechanically-regulated mechanism can help us explore the pathological events in a disease. Here, we reviewed the role of sensory proteins in pathological events that are observed in cardiomyopathy, cancer, respiratory, renal, obesity, genetics, physical injury and bacterial infection. Taken together, sensory proteins are mechanically-activated which assist reception of external physical stimulation and convert into biochemical to trigger intracellular signaling cascade.


2017 ◽  
Author(s):  
Cátia P. Frias ◽  
Tom Bresser ◽  
Lisa Scheefhals ◽  
Hai Yin Hu ◽  
Paul M. P. van Bergen en Henegouwen ◽  
...  

ABSTRACTChanges in inhibitory connections are essential for experience-dependent circuit adaptations. Defects in inhibitory synapses are linked to neurodevelopmental disorders, but the molecular processes underlying inhibitory synapse formation are not well understood. Here we use high resolution two-photon microscopy in organotypic hippocampal slices to examine the signaling pathways induced by the postsynaptic signaling molecule Semaphorin4D (Sema4D) during inhibitory synapse formation. By monitoring changes in individual GFP-labeled presynaptic boutons we found that the primary action of Sema4D is to induce stabilization of presynaptic boutons within tens of minutes. Stabilizing boutons rapidly recruited synaptic vesicles, which was followed by accumulation of postsynaptic gephyrin. Newly formed inhibitory synapses were complete and functional after 24 hours, as determined by electrophysiology and immunohistochemistry. We further showed that Sema4D signaling is regulated by network activity and can induce a local increase in bouton density, suggesting a possible role in circuit adaptation. We further examined the intracellular signaling cascade triggered by Sema4D and found that bouton stabilization occurred through rapid remodeling of actin, and this could be mimicked by the actin-depolymerizing drug Latrunculin B or by reducing ROCK activity. The intracellular signaling cascade required activation of the receptor tyrosine kinase MET, which is a well-known autism risk factor. Our immunohistochemistry data suggests that MET may be localized to presynaptic inhibitory axons. Together, our data yield important insights in the molecular pathway underlying activity-dependent Sema4D-induced synapse formation and reveal a novel role for MET in inhibitory synapses.Significance StatementGABAergic synapses provide the main inhibitory control of neuronal activity in the brain. We make important steps in unraveling the molecular processes that take place when formation of inhibitory synapses is triggered by a specific signaling molecule, Sema4D. We find that this process depends on network activity and involves specific remodeling of the intracellular actin cytoskeleton. We also reveal a previously unknown role for MET in inhibitory synapses. As defects in GABAergic synapses have been implied in many brain disorders, and mutations in MET are strong risk factors for autism, our findings urge for a further investigation of the role of MET at inhibitory synapses.


Sign in / Sign up

Export Citation Format

Share Document