epigenetic remodeling
Recently Published Documents


TOTAL DOCUMENTS

162
(FIVE YEARS 69)

H-INDEX

30
(FIVE YEARS 6)

2022 ◽  
Vol 145 ◽  
pp. 112431
Author(s):  
Hung-Yu Lin ◽  
Hsing-Ju Wu ◽  
Si-Yun Chen ◽  
Ming-Feng Hou ◽  
Chang-Shen Lin ◽  
...  

Forests ◽  
2021 ◽  
Vol 12 (12) ◽  
pp. 1784
Author(s):  
Houda Ben Slama ◽  
Ali Chenari Bouket ◽  
Faizah N. Alenezi ◽  
Zeinab Pourhassan ◽  
Patrycja Golińska ◽  
...  

World population growth and modernization have engendered multiple environmental problems: the propagation of humans and crop diseases and the development of multi-drug-resistant fungi, bacteria and viruses. Thus, a considerable shift towards eco-friendly products has been seen in medicine, pharmacy, agriculture and several other vital sectors. Nowadays, studies on endophytic fungi and their biotechnological potentials are in high demand due to their substantial, cost-effective and eco-friendly contributions in the discovery of an array of secondary metabolites. For this review, we provide a brief overview of plant–endophytic fungi interactions and we also state the history of the discovery of the untapped potentialities of fungal secondary metabolites. Then, we highlight the huge importance of the discovered metabolites and their versatile applications in several vital fields including medicine, pharmacy, agriculture, industry and bioremediation. We then focus on the challenges and on the possible methods and techniques that can be used to help in the discovery of novel secondary metabolites. The latter range from endophytic selection and culture media optimization to more in-depth strategies such as omics, ribosome engineering and epigenetic remodeling.


2021 ◽  
Author(s):  
Adrienne Scott ◽  
Eduard Casas ◽  
Stephanie Ellyse Schneider ◽  
Alison Swearingen ◽  
Courtney Van Den Elzen ◽  
...  

Understanding how cells remember previous mechanical environments to influence their fate, or mechanical memory, informs the design of biomaterials and therapies in medicine. Current regeneration therapies require two-dimensional (2D) cell expansion processes to achieve large cell populations critical for the repair of damaged (e.g. connective and musculoskeletal) tissues. However, the influence of mechanical memory on cell fate following expansion is unknown, and mechanisms defining how physical environments influence the therapeutic potential of cells remain poorly understood. Here, we show that the organization of histone H3 trimethylated at lysine 9 (H3K9me3) and expression of tissue-identifying genes in primary cartilage cells (chondrocytes) transferred to three-dimensional (3D) hydrogels depends on the number of previous population doublings on tissue culture plastic during 2D cell expansion. Decreased levels of H3K9me3 occupying promoters of dedifferentiation genes after the 2D culture were also retained in 3D culture. Suppression of H3K9me3 during expansion of cells isolated from a murine model similarly resulted in the loss of the chondrocyte phenotype and global remodeling of nuclear architecture. In contrast, increasing levels of H3K9me3 through inhibiting H3K9 demethylases partially rescued the chondrogenic nuclear architecture and gene expression, which has important implications for tissue repair therapies, where expansion of large numbers of phenotypically-suitable cells is required. Overall, our findings indicate mechanical memory in primary cells is encoded in the chromatin architecture, which impacts cell fate and the phenotype of expanded cells.


2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. 668-668
Author(s):  
Peter Niimi ◽  
Margarita Meer ◽  
Morgan Levine

Abstract The epigenetic landscape is remodeled with age, bringing about widespread consequences for cell function. With the revolutionary discoveries by Yamanaka and Takahashi, as well as those that built on this work, the transcription factors Oct4, Sox2, KLF4, and C-Myc (OSKM) can be expressed in a variety of cells, including fibroblasts, to make iPSCs. Once cells are reprogrammed, they show an erasure of epigenetic remodeling, suggesting an avenue to reverse aging. It has been recently shown that ectopic expression of three factors, OSK, can restore vision in mouse glaucoma model and reduces epigenetic age. It is not known the path epigenetic remodeling takes or whether all three factors, OSK, are required to remodel the epigenetic landscape. We hypothesize that during reprogramming, cells will reverse along a similar path they took during aging and eventually reverse along that path they took during differentiation. Alternatively, it may also be possible that cells take entirely new paths to reach a state of partial reprogramming or pluripotency. We used DNA methylation and RNA-seq as a multi-omics approach to map the trajectories cells make during aging, differentiation, and reprogramming. In human fibroblasts and hepatocytes, we tested the three-factor OSK mix, as well as pairwise factors OS, OK, and SK and individual Oct4, Sox2, and KLF4 for their effect on cell trajectories. This study provides a dynamic model for epigenetic changes in aging, differentiation, and reprogramming and highlights barriers and bottlenecks throughout the process.


2021 ◽  
Vol 12 ◽  
Author(s):  
Celeste Faia ◽  
Karlie Plaisance-Bonstaff ◽  
Cecilia Vittori ◽  
Dorota Wyczechowska ◽  
Adam Lassak ◽  
...  

Persons living with HIV (PLWH) are at higher risk of developing secondary illnesses than their uninfected counterparts, suggestive of a dysfunctional immune system in these individuals. Upon exposure to pathogens, monocytes undergo epigenetic remodeling that results in either a trained or a tolerant phenotype, characterized by hyper-responsiveness or hypo-responsiveness to secondary stimuli, respectively. We utilized CD14+ monocytes from virally suppressed PLWH and healthy controls for in vitro analysis following polarization of these cells toward a pro-inflammatory monocyte-derived macrophage (MDM) phenotype. We found that in PLWH-derived MDMs, pro-inflammatory signals (TNFA, IL6, IL1B, miR-155-5p, and IDO1) dominate over negative feedback signals (NCOR2, GSN, MSC, BIN1, and miR-146a-5p), favoring an abnormally trained phenotype. The mechanism of this reduction in negative feedback involves the attenuated expression of IKZF1, a transcription factor required for de novo synthesis of RELA during LPS-induced inflammatory responses. Furthermore, restoring IKZF1 expression in PLWH-MDMs partially reinstated expression of negative regulators of inflammation and lowered the expression of pro-inflammatory cytokines. Overall, this mechanism may provide a link between dysfunctional immune responses and susceptibility to co-morbidities in PLWH with low or undetectable viral load.


2021 ◽  
Author(s):  
Nilesh Kumar Sharma ◽  
Sachin C Sarode ◽  
Gargi S Sarode ◽  
Shankargouda Patil

Accumulating evidence suggests the role of cellular components in achieving antitumor to protumor microenvironments. Among the various types of cells within the tumor niche, the state of CD8+ T cells apparently changes from cytotoxic T effector cells and memory T cells to exhausted CD8+ T cells. These changes in the phenotype of CD8+ T cells promote the protumor microenvironment. Recently, comprehensive experimental data delineated the role of thymocyte selection-associated high-mobility group-box protein (TOX), which regulates the transcriptional process and epigenetic remodeling, with implications in tumor and chronic viral infections. This perspective summarizes the molecular mechanisms that link CD8+ T cells, TOX, and transcriptional and epigenetic reprogramming as well as future directions for determining new avenues of cancer therapeutics.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2410-2410
Author(s):  
Edd C Ricker ◽  
Brian Estrella ◽  
Manuel A Pazos ◽  
Jennifer E. Amengual

Abstract Epigenetic remodeling is essential for the proper differentiation and function of germinal center (GC) B cells. Competing actions of histone acetyltransferases (HATs), such as CREBBP and EP300, and inhibitory histone deacetylases (HDACs) and methyltransferases (HMTs), such as EZH2, modulate the epigenomic program of GC B cells and control key activating and immunogenic processes, such as BCR and CD40 signaling, antigen processing and presentation, and cell cycle regulation. In line with the critical importance of epigenetic regulation of GC B cell function, inactivating mutations in CREBBPor EP300and gain-of-function mutations in EZH2 have been identified in 39% and 21% of GC B-cell like diffuse large B-cell lymphoma (GC-DLBCL), respectively. Strategies to target epigenetic dysfunction in GC-DLBCL have largely focused on indirectly restoring the activity of HATs through the use of histone deacetylase (HDAC) inhibitors, such as belinostat, and by directly inhibiting EZH2 with tazemetostat. Despite showing great promise in preclinical studies, single agent therapies with HDAC or EZH2 inhibitors have shown only modest efficacy in the clinic. Recently, our group demonstrated synergistic effects of dual therapy with EZH2 and HDAC inhibitors in inducing lethality of GC-DLBCL lines. As combination therapies begin to be tested in clinical trials, whether dual inhibition of EZH2 and HDAC is able to restore immunogenic features of GC-DLBCL and lead to enhanced T cell-mediated killing in vivo remains unknown. Here, we utilize transcriptomic and flow cytometric methods to assess whether epigenetic remodeling with tazemetostat (EZH2 inhibitor) and belinostat (HDAC inhibitor) can alter the immunogenicity of GC-DLBCL. We find that combination treatment with both tazemetostat and belinostat promotes an antigen processing and presentation program in GC-DLBCL lines and results in significant increases in MHC-I and MHC-II surface expression. This effect was only modestly appreciated with single agent treatment and required at least 4-7 days post-treatment to become apparent. Importantly, the increased MHC expression in response to combination therapy was not dependent on EZH2 or CREBBPmutation status, suggesting broad applications of dual therapy in patients with diverse mutation burdens. Furthermore, we found that combination treatment also altered surface expression of costimulatory molecules, such as CD80 and CD86, suggesting that tazemetostat and belinostat can modulate interactions with T cells in a multifaceted manner. These findings thus uncover that dual targeting of EZH2 and HDAC with tazemetostat and belinostat promotes antigen presentation pathways in GC-DLBCL and suggests that dual therapies could restore immunogenicity in GC-DLBCL and enhance immune-mediated killing of tumor cells. This work was funded, in part, by the ASH HONORS Award Summer Program. Figure 1 Figure 1. Disclosures Amengual: Seagen: Consultancy; Appia Pharmaceuticals: Research Funding; Daiichi Sankyo, Inc: Consultancy; Epizyme, Inc.: Speakers Bureau.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 677-677
Author(s):  
Kevin A. Nuno ◽  
Armon Azizi ◽  
Thomas Koehnke ◽  
Asiri Ediriwickrema ◽  
M. Ryan Corces ◽  
...  

Abstract Introduction: Acute myeloid leukemia (AML) is associated with a poor prognosis and high rates of relapse despite aggressive treatments including high dose chemotherapy. To understand the clonal dynamics and genetic evolution of relapsed AML, we analyzed a cohort of 142 previously published genotyped and paired diagnosis-relapse AML samples. 40% of queried cases exhibited no major changes in somatic mutations upon relapse, and genetically stable clonal structure correlated with increased relapse probability. Thus, we hypothesized epigenetic reprogramming plays a role in these cases and AML relapse in general. Here, we examine the epigenetic landscape of relapsed AML and characterize the cis and trans regulatory elements that correlate with AML relapse in the presence or absence of genetic evolution. Methods: We identified 27 viable cryopreserved paired diagnosis and relapse samples from the same patient treated at Stanford with high-dose chemotherapy. Leukemic blasts and (when possible) leukemia stem cell (LSC) enriched populations were FACS purified and prepared for genotyping with a myeloid malignancy targeted sequencing panel as well as ATAC-seq for chromatin accessibility profiling. Single-cell ATAC-seq was further performed on select samples to investigate regulatory reprogramming in cell subpopulations. We then performed integrative analysis to uncover the interplay between genetic lesions, epigenetic regulatory programs, and gene accessibility in relapsed AML. Results: Genotyping analysis of these AML specimens revealed that 40% of samples exhibited no changes in AML-related genetic alterations upon relapse (hereby referred to as "stable" samples). Chromatin accessibility analysis revealed these stable samples had a distinct epigenetic signature, modulating similar gene accessibility programs and sharing enhancer loci that become accessible across all stable relapse samples. These sequences included genes involved in chromatin organization and compaction, as well as those involved in transcriptional control of hematopoietic differentiation and myeloid cell maturation. We also observed several regulatory signatures present at relapse specific for AML subtypes, including NPM1/FLT3 double mutant AML and those with mutations in transcription factors such as CEBPA or RUNX1. We then performed single-cell ATAC-seq on genetically stable samples to further characterize the sub-clonal epigenetic dynamics between the diagnosis and relapse cells. Several samples exhibited regulatory heterogeneity in multiple cell subpopulations that changed significantly at relapse. One subpopulation of interest was characterized by increased GATA and RUNX family transcription factor motif accessibility at relapse, indicating a shift toward a less differentiated progenitor cell phenotype. In addition, we identified a subpopulation of cells at diagnosis that were epigenetically similar to the major epigenetic states present at relapse, indicating that selection for specific epigenetic subclones may occur in AML patients during therapy in the absence of additional genetic lesions. Finally, given the critical role of LSCs in AML pathogenesis and their possible role as a reservoir for AML relapse, we analyzed LSC-enriched subpopulations in a subset of our cohort. ATAC-seq analysis indicated these cells shared several LSC-specific epigenetic features between samples, are distinguished from leukemia blasts by distinct regulatory programs, and undergo epigenetic remodeling between initial diagnosis and relapse. Gene accessibility analysis also revealed a shared LSC gene expression signature that also shifted at relapse. These data indicate a specific, distinct epigenomic signature for LSC enriched cell populations, and that epigenetic evolution at relapse occurs intracellularly, rather than reflecting heterogeneity in cellular subpopulations upon AML relapse. Conclusion: This study reveals that epigenetic remodeling in the absence of genetic evolution is a mechanism through which AML relapse occurs. We show that chromatin reorganization of genes and regulatory sequences occurs in these AML cells, leading to a permissive cell state that might be resistant to conventional treatment. Ongoing work includes dissecting the subclonal structure of these AML cells and identifying the relationship between gene regulatory networks that contribute to relapse. Disclosures Ediriwickrema: Nanosive SAS: Patents & Royalties. Majeti: BeyondSpring Inc.: Membership on an entity's Board of Directors or advisory committees; CircBio Inc.: Membership on an entity's Board of Directors or advisory committees; Kodikaz Therapeutic Solutions Inc.: Membership on an entity's Board of Directors or advisory committees; Coherus Biosciences: Membership on an entity's Board of Directors or advisory committees; Acuta Capital Partners: Consultancy; Gilead: Patents & Royalties: inventor on a number of patents related to CD47 cancer immunotherapy licensed to Gilead Sciences, Inc..


Sign in / Sign up

Export Citation Format

Share Document