scispace - formally typeset
Search or ask a question

How do histone crotonylation reinforce HIV latency? 


Best insight from top research papers

Histone crotonylation reinforces HIV latency by reprogramming the local chromatin at the HIV long-terminal repeat (LTR) through increased histone acetylation and reduced histone methylation . This epigenetic modification of the HIV LTR is regulated by the expression of the crotonyl-CoA-producing enzyme acyl-CoA synthetase short-chain family member 2 (ACSS2) . Induction of ACSS2 leads to increased histone crotonylation, which in turn promotes the reactivation of latent HIV . Inhibition or knockdown of ACSS2 diminishes histone crotonylation-induced HIV replication and reactivation . Furthermore, the induction of ACSS2 is highly synergistic with protein kinase C agonists or histone deacetylase inhibitors in reactivating latent HIV . This study identifies histone lysine crotonylation as a novel epigenetic regulator for HIV transcription that can be targeted for HIV eradication .

Answers from top 5 papers

More filters
Papers (5)Insight
The provided paper does not mention histone crotonylation or its role in reinforcing HIV latency.
The provided paper does not mention anything about histone crotonylation reinforcing HIV latency.
Histone crotonylation is a regulator of HIV latency. Induction of histone crotonylation through increased expression of the enzyme ACSS2 reprograms the chromatin at the HIV LTR, leading to increased histone acetylation and reduced histone methylation, which in turn promotes HIV replication and reactivation.
The provided paper does not specifically mention how histone crotonylation reinforces HIV latency.
The provided paper does not mention histone crotonylation or its role in reinforcing HIV latency.

Related Questions

What are histone modifications?5 answersHistone modifications are chemical changes to the amino acid residues of histone proteins that regulate gene expression without altering the DNA sequence. These modifications include methylation, acetylation, phosphorylation, ubiquitination, and many others. They form a "histone code" that can be used as an epigenetic marker and play a crucial role in various cellular processes such as transcriptional regulation, DNA repair, and chromosome segregation. Histone-modifying enzymes, such as histone acetyltransferases (HATs), histone deacetylases (HDACs), and histone methyltransferases (KMTs), add or remove these modifications, thereby altering the functional profile of the epigenome. Understanding the role of histone post-translational modifications is important for preventing and treating human diseases, including cancer.
Is the turnover of histone methylation slower than the turnover of histone acetylation?5 answersHistone methylation turnover is slower than histone acetylation turnover. The turnover rate of histone acetylation is generally faster than that of histone methylation. The turnover of histone acetylation varies depending on the histone, the modified residue, and neighboring modifications. In contrast, histone methylation, such as H3 trimethyl K9 and K27, is slow to be established on new histones and slow to disappear from old histones. The turnover rates of transcription-associated marks, such as K4me3 and K36me3, are faster than histone methylation. These findings suggest that histone acetylation has a faster turnover rate compared to histone methylation.
Are histone tail modifications heritable?4 answers
Can histone modifications increase gene expression?11 answers
How do you get rid of histones?3 answers
Is histone modification random?3 answers

See what other people are reading

What are the interactions between the Lysophospholipids and CD103?
7 answers
Lysophospholipids (LPLs) and CD103 interact within the immune and tumor microenvironments through complex mechanisms that influence cell signaling, adhesion, and immune responses. LPLs such as lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P) are bioactive molecules that play significant roles in cellular processes, including immune cell function and tumor pathogenesis. These molecules exert their effects by binding to specific receptors on cell surfaces, leading to activation and proliferation of various cell types, including T cells. In the context of cancer, LPLs contribute to the modulation of the tumor microenvironment by affecting cell proliferation, survival, motility, angiogenesis, and immune system interactions. CD103, an integrin expressed on tumor-infiltrating lymphocytes (TILs), interacts with E-cadherin on epithelial tumor cells, triggering polarized exocytosis of cytotoxic granules that lead to tumor cell lysis. This interaction is crucial for the activation and effector functions of TILs, highlighting the importance of CD103 in mediating immune responses against tumors. Furthermore, CD103 has been identified as a potential target for preventing or treating allograft rejection, indicating its significant role in modulating immune responses. The interaction between LPLs and CD103 can be inferred from their roles in the immune system and tumor microenvironment. LPLs, through their receptors, can influence the function of CD8 T cells, which express CD103, by promoting tolerogenic states and affecting metabolic reprogramming and differentiation. This suggests that LPLs may indirectly influence CD103 function by modulating the activity and state of CD8 T cells within the tumor microenvironment. In summary, while direct interactions between LPLs and CD103 are not explicitly detailed in the provided contexts, the roles of LPLs in modulating immune cell functions and the critical function of CD103 in T-cell–mediated cancer cell killing suggest a complex interplay between these molecules in regulating immune responses and tumor dynamics.
What are the implications of BRCA1 mutation on the efficacy of personalized therapeutic interventions for breast cancer?
5 answers
BRCA1 mutations play a significant role in the efficacy of personalized therapeutic interventions for breast cancer. Mutations in BRCA1 lead to DNA repair defects, making tumors more vulnerable to therapies like Poly (ADP-ribose) polymerase inhibitors (PARPi). These mutations result in homologous recombination repair deficiencies, affecting responses to platinum salt therapies and PARP inhibitors. However, the efficacy of these treatments can be impacted by the development of resistance due to interconnected DNA repair pathways compensating for each other. Additionally, BRCA1 promoter methylation can lead to poor outcomes and adaptive chemoresistance, altering therapeutic responses in TNBC and ovarian cancers. Understanding the implications of BRCA1 mutations is crucial for tailoring effective treatment strategies in breast cancer patients.
What are the most effective chondroprotectors?
4 answers
The most effective chondroprotectors include collagen hydrolysates, sulfated glucosamine, chondroitin sulfate, glucosamine sulfate or hydrochloride, hyaluronic acid, and plant-based extracts like Kalopanax pictus Castor-Aralia, Hericium erinaceus, and Astragalus membranaceus. Additionally, marine-derived HDAC inhibitor panobinostat has shown promising chondroprotective effects by attenuating cartilage degeneration and regulating key proteins in chondrocytes. Other potential chondroprotective agents include polyunsaturated fatty acids, S-adenosylmethionine, avocado, soybean unsaponifiable fractions, methylsulfonylmethane, vitamins C, D, and E, intra-articular hyaluronic acid injections, and platelet-rich plasma. These substances have demonstrated efficacy in preserving articular cartilage, reducing inflammation, and promoting cartilage health, making them valuable options for managing osteoarthritis.
How to identify promoter enhancer interaction?
10 answers
Identifying promoter-enhancer interactions (PEIs) is crucial for understanding the regulatory mechanisms that control gene expression. Recent advancements in both experimental and computational methods have significantly improved our ability to detect these interactions. High-throughput chromosome conformation capture assays, such as Hi-C, have been instrumental in studying basic chromatin architecture and revealing the physical proximity between enhancers and promoters. Additionally, technologies like STARR-seq quantify promoter and enhancer activity, while CRISPRi helps identify enhancer-promoter compatibility. On the computational front, machine learning models have been widely adopted for predicting PEIs. These models often require a combination of functional genomic and epigenomic features as input. For instance, the HARD model uses a minimal set of features, including H3K27ac, ATAC-seq, RAD21, and distance metrics, to predict EPIs with high accuracy. Similarly, EPIXplorer integrates multiple predictive algorithms and supports various types of 3D contact and multi-omics data, offering a user-friendly platform for EPI prediction. Another approach, StackEPI, leverages a stacking ensemble learning strategy to efficiently predict cell line-specific EPIs. Emerging methods also focus on the role of DNA-binding proteins and transcription factors in mediating EPIs. The PENGUIN approach, for example, integrates chromatin interactions with tissue-specific protein-protein interaction networks to identify and analyze EPI networks in the context of human diseases. Furthermore, novel computational techniques like the digital Hi-C assay combined with transformer algorithms allow for the prediction of long-distance genomic interactions based on limited sequence data. Innovative models such as TransEPI and EPI-DLMH have also been developed, utilizing deep learning to capture long-range dependencies and interactions between enhancers and promoters. These models demonstrate superior performance in predicting EPIs across different cell lines and conditions, highlighting the potential of computational approaches to complement experimental methods in identifying and understanding EPIs.
How genetics involved in aspirin responsiveness?
5 answers
Genetic factors play a crucial role in aspirin responsiveness. Studies have shown that genetic variation in bitter taste receptors (T2Rs) can impact the immune response in the upper airway. Additionally, genetic variants in adenosine kinase have been associated with aspirin response, particularly affecting purine metabolism pathways. The Aspirin Response Signature (ARS) score, which is based on prothrombotic gene transcripts, can help individualize antiplatelet therapy, with higher scores correlating with lower platelet function and increased risk of adverse cardiovascular events. These genetic insights into aspirin responsiveness pave the way for personalized treatment strategies in cardiovascular disease management.
What molecular mechanisms are involved in the differentiation of eye tissues during embryonic development?
5 answers
During embryonic development, the differentiation of eye tissues involves intricate molecular mechanisms regulated by epigenetic factors. These mechanisms include the dynamic changes in chromatin structure, such as alterations in DNA methylation patterns and histone modifications, which impact gene expression during tissue differentiation. Specifically, the core-promoter variations play a crucial role in defining transcriptional output during cell fate transitions in the developing eye. Furthermore, the activation of specific transcription factors, like Pax6, and the presence of cis-regulatory DNA motifs, such as AP-1, Ets, and Maf sites, are essential for orchestrating the differentiation of eye tissues, including lens fibers and epithelium. These molecular events contribute to the precise spatio-temporal control of gene expression, lineage commitment, and fate determination in the developing vertebrate eye.
What are some recently identified potential drug targets for serous ovarian cancer?
5 answers
Recent research has identified several potential drug targets for serous ovarian cancer. These targets include amplified genes such as CCNE1, PAX8, URI1, PRKCI, and FAL1, which have shown promise as therapeutic targets. Additionally, mutations in KRAS and NRAS have been linked to sensitivity or resistance to MEK inhibitors in low-grade serous ovarian carcinoma, suggesting these genes as potential targets. Epigenomic profiling has highlighted SNS-032 and inhibitors BIX-01294 and UNC0646 as therapeutic candidates for high-grade serous carcinoma. Furthermore, somatic mutations in genes like PARP have been identified in high-grade serous ovarian cancer patients, indicating PARP inhibitors as potential targeted therapies. These findings underscore the diverse array of potential drug targets that are being explored for serous ovarian cancer treatment.
How to reduce the methylglyoxal by genetic engineering in mammalian cells??
5 answers
To reduce methylglyoxal (MGO) levels in mammalian cells through genetic engineering, several strategies can be employed. One approach involves modulating the cysteine/glutathione metabolic pathways or regulating glucose consumption to control acidic species like MGO. Additionally, targeting the glyoxalase system, which detoxifies MGO to D-lactate, can be crucial in reducing MGO-associated damage, especially in conditions like diabetes. Studies have shown that in the absence of Glyoxalase I (Glo1), aldose-keto reductases (AKR) can effectively compensate to prevent MGO accumulation, highlighting a potential pathway for genetic manipulation to mitigate MGO levels in diabetic conditions. Furthermore, manipulating the translation of Notch1 receptor mRNA through methylglyoxal-mediated inhibition of GAPDH enzymatic activity can impact Notch signaling and NPC fate decisions, offering another genetic engineering avenue to regulate MGO levels in mammalian cells.
What is banana pseudoephedrine?
4 answers
Banana pseudoephedrine does not refer to a specific product or term in the contexts provided. Pseudoephedrine is a drug commonly used as a nasal decongestant and for various medical purposes like treating cold symptoms, asthma, and bronchitis. Studies have shown that pseudoephedrine can impact T-cell activation events, inhibiting certain gene transcription and transcriptional activities of key factors. Additionally, pseudoephedrine is known to cause blood vessel constriction, aiding in reducing nasal airflow resistance. Research has also explored the effects of pseudoephedrine on exercise performance, showing improvements in running trials without reported side effects. Furthermore, high doses of pseudoephedrine have been linked to sperm abnormalities, decreased sperm count, and increased apoptotic activity in rat testes.
Inhibition of yH2AX phosphorylation?
4 answers
Inhibition of γ-H2AX phosphorylation can have varying effects depending on the context. Studies have shown that during herpes simplex virus (HSV) infections, inhibition of ATM kinase activity can prevent HSV-stimulated H2AX phosphorylation, with minor effects on DNA replication and virus yield. Additionally, in response to ionizing radiation, inhibition of γ-H2AX has been linked to aberrant DNA repair and increased radiosensitivity in prostate cancer cells. Furthermore, the phosphorylation of H2AX after UV irradiation is triggered by DNA repair intermediates and is primarily mediated by the ATR kinase, suggesting a role for ATR in this process. These findings highlight the complex interplay between H2AX phosphorylation and various cellular processes, emphasizing the importance of understanding the specific context in which inhibition occurs.
What does IL-2 do with T cells?
5 answers
IL-2 plays a crucial role in regulating T cell responses. It can promote T cell proliferation, differentiation into various subsets like Th1, Th2, regulatory T, and cytotoxic T cells, and inhibit Th17 differentiation. IL-2 can induce T cell exhaustion within tumor microenvironments by activating STAT5, leading to the upregulation of inhibitory receptors and downregulation of effector-molecule production, ultimately rendering T cells dysfunctional. Additionally, IL-2 has been shown to enhance antitumor activity by acting as a partial agonist that avoids the terminal differentiation of T cells. Furthermore, re-engineered IL-2 therapeutics are being developed to selectively stimulate specific T cell subsets, such as effector T cells or regulatory T cells, depending on the therapeutic goal for cancer or autoimmune diseases.