Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
1,206
result(s) for
"631/250/2502"
Sort by:
The regulation of immune tolerance by FOXP3
by
Barbi, Joseph
,
Pan, Fan
,
Lu, Ling
in
631/250/1619/554/1898/1271
,
631/250/2502
,
631/250/2502/2170
2017
Key Points
Forkhead box protein P3 (FOXP3) is a crucial regulator of regulatory T (T
reg
) cell gene expression that is responsible for much of the suppressive potential displayed by these cells.
The regulation of
FOXP3
expression in T
reg
cells occurs through the concerted action of transcription factors and extensive epigenetic control mechanisms; furthermore, post-translational modifications are also capable of modulating FOXP3 function.
These several layers of FOXP3 control are responsive to positive and negative regulation by factors in the tissue environment, including cytokines, inflammatory mediators and metabolic factors.
Modulating FOXP3 expression and T
reg
cell function by targeting newly discovered regulatory nodes may lead to the development of new immunotherapies for cancer and autoimmune diseases.
This Review considers how forkhead box protein P3 (FOXP3) — the key transcription factor of regulatory T (T
reg
) cells — is regulated both at the transcriptional level and through post-translational modifications. The authors explain how FOXP3 interacts with other molecules to induce and maintain T
reg
cell populations, and they discuss the potential of therapeutically targeting FOXP3 in the context of human disease.
The proper restraint of the destructive potential of the immune system is essential for maintaining health. Regulatory T (T
reg
) cells ensure immune homeostasis through their defining ability to suppress the activation and function of other leukocytes. The expression of the transcription factor forkhead box protein P3 (FOXP3) is a well-recognized characteristic of T
reg
cells, and FOXP3 is centrally involved in the establishment and maintenance of the T
reg
cell phenotype. In this Review, we summarize how the expression and activity of FOXP3 are regulated across multiple layers by diverse factors. The therapeutic implications of these topics for cancer and autoimmunity are also discussed.
Journal Article
Sexual-dimorphism in human immune system aging
2020
Differences in immune function and responses contribute to health- and life-span disparities between sexes. However, the role of sex in immune system aging is not well understood. Here, we characterize peripheral blood mononuclear cells from 172 healthy adults 22–93 years of age using ATAC-seq, RNA-seq, and flow cytometry. These data reveal a shared epigenomic signature of aging including declining naïve T cell and increasing monocyte and cytotoxic cell functions. These changes are greater in magnitude in men and accompanied by a male-specific decline in B-cell specific loci. Age-related epigenomic changes first spike around late-thirties with similar timing and magnitude between sexes, whereas the second spike is earlier and stronger in men. Unexpectedly, genomic differences between sexes increase after age 65, with men having higher innate and pro-inflammatory activity and lower adaptive activity. Impact of age and sex on immune phenotypes can be visualized at
https://immune-aging.jax.org
to provide insights into future studies.
Whether the immune system aging differs between men and women is barely known. Here the authors characterize gene expression, chromatin state and immune subset composition in the blood of healthy humans 22 to 93 years of age, uncovering shared as well as sex-unique alterations, and create a web resource to interactively explore the data.
Journal Article
Epigenetic control of CD8+ T cell differentiation
by
Henning, Amanda N
,
Roychoudhuri, Rahul
,
Restifo, Nicholas P
in
Cancer
,
CD8 antigen
,
Cell differentiation
2018
Upon stimulation, small numbers of naive CD8+ T cells proliferate and differentiate into a variety of memory and effector cell types. CD8+ T cells can persist for years and kill tumour cells and virally infected cells. The functional and phenotypic changes that occur during CD8+ T cell differentiation are well characterized, but the epigenetic states that underlie these changes are incompletely understood. Here, we review the epigenetic processes that direct CD8+ T cell differentiation and function. We focus on epigenetic modification of DNA and associated histones at genes and their regulatory elements. We also describe structural changes in chromatin organization that affect gene expression. Finally, we examine the translational potential of epigenetic interventions to improve CD8+ T cell function in individuals with chronic infections and cancer.
Journal Article
ImmGen report: sexual dimorphism in the immune system transcriptome
2019
Sexual dimorphism in the mammalian immune system is manifested as more frequent and severe infectious diseases in males and, on the other hand, higher rates of autoimmune disease in females, yet insights underlying those differences are still lacking. Here we characterize sex differences in the immune system by RNA and ATAC sequence profiling of untreated and interferon-induced immune cell types in male and female mice. We detect very few differentially expressed genes between male and female immune cells except in macrophages from three different tissues. Accordingly, very few genomic regions display differences in accessibility between sexes. Transcriptional sexual dimorphism in macrophages is mediated by genes of innate immune pathways, and increases after interferon stimulation. Thus, the stronger immune response of females may be due to more activated innate immune pathways prior to pathogen invasion.
Sexual dimorphism is observed frequently in immune disorders, but the underlying insights are still unclear. Here the authors analyze transcriptome and epigenome changes induced by interferon in various mouse immune cell types, and find only a restricted set of sexual dimorphism genes in innate immunity and macrophages.
Journal Article
Novel specialized cell state and spatial compartments within the germinal center
by
Kennedy, Domenick E.
,
McLean, Kaitlin C.
,
Maienschein-Cline, Mark
in
631/250/1619/40
,
631/250/2152/2153/1982
,
631/250/2502/2170
2020
Within germinal centers (GCs), complex and highly orchestrated molecular programs must balance proliferation, somatic hypermutation and selection to both provide effective humoral immunity and to protect against genomic instability and neoplastic transformation. In contrast to this complexity, GC B cells are canonically divided into two principal populations, dark zone (DZ) and light zone (LZ) cells. We now demonstrate that, following selection in the LZ, B cells migrated to specialized sites within the canonical DZ that contained tingible body macrophages and were sites of ongoing cell division. Proliferating DZ (DZp) cells then transited into the larger DZ to become differentiating DZ (DZd) cells before re-entering the LZ. Multidimensional analysis revealed distinct molecular programs in each population commensurate with observed compartmentalization of noncompatible functions. These data provide a new three-cell population model that both orders critical GC functions and reveals essential molecular programs of humoral adaptive immunity.
Germinal centers are typically divided into dark and light zones. Clark and colleagues identify ‘gray zone’ cyclin B1
+
B cell clusters as sites of ongoing cell proliferation, and these cells are distinct from dark zone B cells that undergo AID-dependent somatic hypermutation. This separation of function safeguards B cells undergoing DNA replication against potential mutagenic events that could result in neoplastic transformation.
Journal Article
Chromatin state and BACH2 control TH17 cell functional heterogeneity
2024
We performed transcriptional and chromatin accessibility profiling of T
H
17 cells to distinguish the pathways that regulate pathogenic versus non-pathogenic T
H
17 cell subsets. We show that T
H
17 cell functional heterogeneity is linked to distinct regulatory programs that are shared between T
H
17 cells and other CD4
+
T cell states. BACH2 was identified as a key regulator of T
H
17 cell-mediated autoimmunity.
Journal Article
Hierarchical regulation of the resting and activated T cell epigenome by major transcription factor families
by
Rudensky, Alexander Y
,
Leslie, Christina S
,
Walker, Sarah K
in
Adaptive immunity
,
CD4 antigen
,
CD8 antigen
2022
T cell activation, a key early event in the adaptive immune response, is subject to elaborate transcriptional control. In the present study, we examined how the activities of eight major transcription factor (TF) families are integrated to shape the epigenome of naive and activated CD4 and CD8 T cells. By leveraging extensive polymorphisms in evolutionarily divergent mice, we identified the ‘heavy lifters’ positively influencing chromatin accessibility. Members of Ets, Runx and TCF/Lef TF families occupied the vast majority of accessible chromatin regions, acting as ‘housekeepers’, ‘universal amplifiers’ and ‘placeholders’, respectively, at sites that maintained or gained accessibility upon T cell activation. In addition, a small subset of strongly induced immune response genes displayed a noncanonical TF recruitment pattern. Our study provides a key resource and foundation for the understanding of transcriptional and epigenetic regulation in T cells and offers a new perspective on the hierarchical interactions between critical TFs.Zhong et al. utilize B6/Cast F1 hybrid mice to examine transcriptional regulation of T cell gene expression upon activation induced by viral challenge. They describe gene accessibility changes that lead to differential gene expression and report a hierarchy of transcription factor families that mediate the chromatin dynamics.
Journal Article
B cell activation and plasma cell differentiation are inhibited by de novo DNA methylation
2018
B cells provide humoral immunity by differentiating into antibody-secreting plasma cells, a process that requires cellular division and is linked to DNA hypomethylation. Conversely, little is known about how de novo deposition of DNA methylation affects B cell fate and function. Here we show that genetic deletion of the de novo DNA methyltransferases
Dnmt3a
and
Dnmt3b
(Dnmt3-deficient) in mouse B cells results in normal B cell development and maturation, but increased cell activation and expansion of the germinal center B cell and plasma cell populations upon immunization. Gene expression is mostly unaltered in naive and germinal center B cells, but dysregulated in Dnmt3-deficient plasma cells. Differences in gene expression are proximal to Dnmt3-dependent DNA methylation and chromatin changes, both of which coincide with E2A and PU.1-IRF composite-binding motifs. Thus, de novo DNA methylation limits B cell activation, represses the plasma cell chromatin state, and regulates plasma cell differentiation.
DNA methylation is known to contribute to B cell differentiation, but de novo methylation has not been studied in this context. Here the authors use a conditional Dnmt3a/b knockout mouse to map the function of de novo DNA methylation in B cell differentiation and the development of humoral immunity.
Journal Article
Autocrine vitamin D signaling switches off pro-inflammatory programs of TH1 cells
by
Kazemian Majid
,
Lombardi Giovanna
,
Teague, Heather
in
Autocrine signalling
,
Bronchus
,
c-Jun protein
2022
The molecular mechanisms governing orderly shutdown and retraction of CD4+ type 1 helper T (TH1) cell responses remain poorly understood. Here we show that complement triggers contraction of TH1 responses by inducing intrinsic expression of the vitamin D (VitD) receptor and the VitD-activating enzyme CYP27B1, permitting T cells to both activate and respond to VitD. VitD then initiated the transition from pro-inflammatory interferon-γ+ TH1 cells to suppressive interleukin-10+ cells. This process was primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating super-enhancers and recruiting several transcription factors, notably c-JUN, STAT3 and BACH2, which together with VitD receptor shaped the transcriptional response to VitD. Accordingly, VitD did not induce interleukin-10 expression in cells with dysfunctional BACH2 or STAT3. Bronchoalveolar lavage fluid CD4+ T cells of patients with COVID-19 were TH1-skewed and showed de-repression of genes downregulated by VitD, from either lack of substrate (VitD deficiency) and/or abnormal regulation of this system.During homeostasis TH1 cells activate a cell-intrinsic inflammatory shutdown program and shift to IL-10 production. Chauss et al. find that this TH1 homeostatic program is dependent on vitamin D signaling and is disrupted in severe COVID-19.
Journal Article
Epigenetic regulation of T cell exhaustion
by
Daniel, Bence
,
Belk, Julia A
,
Satpathy, Ansuman T
in
Autoimmunity
,
Conformation
,
DNA methylation
2022
Chronic antigen stimulation during viral infections and cancer can lead to T cell exhaustion, which is characterized by reduced effector function and proliferation, and the expression of inhibitory immune checkpoint receptors. Recent studies have demonstrated that T cell exhaustion results in wholescale epigenetic remodeling that confers phenotypic stability to these cells and prevents T cell reinvigoration by checkpoint blockade. Here, we review foundational technologies to profile the epigenome at multiple scales, including mapping the locations of transcription factors and histone modifications, DNA methylation and three-dimensional genome conformation. We discuss how these technologies have elucidated the development and epigenetic regulation of exhausted T cells and functional implications across viral infection, cancer, autoimmunity and engineered T cell therapies. Finally, we cover emerging multi-omic and genome engineering technologies, current and upcoming opportunities to apply these to T cell exhaustion, and therapeutic opportunities for T cell engineering in the clinic.Satpathy and colleagues review the epigenetic underpinnings that result in T cell exhaustion.
Journal Article