Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 384
Filter
1.
J Hematol Oncol ; 17(1): 91, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380002

ABSTRACT

BACKGROUND: The epigenetic factors KAT6A (MOZ/MYST3) and KMT2A (MLL/MLL1) interact in normal hematopoiesis to regulate progenitors' self-renewal. Both proteins are recurrently translocated in AML, leading to impairment of critical differentiation pathways in these malignant cells. We evaluated the potential of different KAT6A therapeutic targeting strategies to alter the growth of KAT6A and KMT2A rearranged AMLs. METHODS: We investigated the action and potential mechanisms of the first-in-class KAT6A inhibitor, WM-1119 in KAT6A and KMT2A rearranged (KAT6Ar and KMT2Ar) AML using cellular (flow cytometry, colony assays, cell growth) and molecular (shRNA knock-down, CRISPR knock-out, bulk and single-cell RNA-seq, ChIP-seq) assays. We also used two novel genetic murine KAT6A models combined with the most common KMT2Ar AML, KMT2A::MLLT3 AML. In these murine models, the catalytic activity of KAT6A, or the whole protein, can be conditionally abrogated or deleted. These models allowed us to compare the effects of specific KAT6A KAT activity inhibition with the complete deletion of the whole protein. Finally, we also tested these therapeutic approaches on human AML cell lines and primary patient AMLs. RESULTS: We found that WM-1119 completely abrogated the proliferative and clonogenic potential of KAT6Ar cells in vitro. WM-1119 treatment was associated with a dramatic increase in myeloid differentiation program. The treatment also decreased stemness and leukemia pathways at the transcriptome level and led to loss of binding of the fusion protein at critical regulators of these pathways. In contrast, our pharmacologic and genetic results indicate that the catalytic activity of KAT6A plays a more limited role in KMT2Ar leukemogenicity, while targeting the whole KAT6A protein dramatically affects leukemic potential in murine KMT2A::MLLT3 AML. CONCLUSION: Our study indicates that inhibiting KAT6A KAT activity holds compelling promise for KAT6Ar AML patients. In contrast, targeted degradation of KAT6A, and not just its catalytic activity, may represent a more appropriate therapeutic approach for KMT2Ar AMLs.


Subject(s)
Histone Acetyltransferases , Histone-Lysine N-Methyltransferase , Leukemia, Myeloid, Acute , Myeloid-Lymphoid Leukemia Protein , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Animals , Myeloid-Lymphoid Leukemia Protein/genetics , Humans , Mice , Histone-Lysine N-Methyltransferase/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/antagonists & inhibitors , Gene Rearrangement , Cell Line, Tumor
2.
Lett Appl Microbiol ; 77(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39257244

ABSTRACT

Histone acetyltransferase inhibitors (HATi) are mechanism-based inhibitors that show promise in the treatment of several illnesses, including diabetes, hyperlipidemia, cancer, and Alzheimer's disease. The work emphasizes the significance of HATi as a possible treatment strategy against Candida species biofilms. Here, in this study, we found that combining a HATi, anacardic acid (AA), and quercetin, a known flavonoid, significantly prevented biofilm formation by C. tropicalis. We further show that C. tropicalis exhibited a considerable downregulation of drug-resistance gene expression (CDR1 and MDR1) when co-administrated. Additionally, in silico studies revealed that the AA interacts strongly with a histone acetyltransferase, Rtt109, which may account for the observed biofilm inhibitory effect. In conclusion, the study illustrates how HATi may be used to potentiate the inhibitory action of phytoactives or antifungals against drug-resistant yeast infections.


Subject(s)
Anacardic Acids , Antifungal Agents , Biofilms , Candida tropicalis , Drug Synergism , Histone Acetyltransferases , Quercetin , Candida tropicalis/drug effects , Quercetin/pharmacology , Biofilms/drug effects , Antifungal Agents/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Anacardic Acids/pharmacology , Drug Resistance, Fungal , Microbial Sensitivity Tests , Enzyme Inhibitors/pharmacology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/antagonists & inhibitors
3.
Cancer Lett ; 604: 217269, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39326554

ABSTRACT

Glioblastoma (GBM) is a fatal adult brain tumor with an extremely poor prognosis. GBM poses significant challenges for targeted therapies due to its intra- and inter-tumoral heterogeneity, a highly immunosuppressive microenvironment, diffuse infiltration into normal brain parenchyma, protection by the blood-brain barrier and acquisition of therapeutic resistance. Recent studies have implicated epigenetic modifiers as key players driving tumorigenesis, resistance, and progression of GBM. While the vast majority of GBM research on epigenetic modifiers thus far has focused predominantly on elucidating the functional roles and targeting of DNA methyltransferases and histone deacetylases, emerging evidence indicates that histone acetyltransferases (HATs) also play a key role in mediating plasticity and therapeutic resistance in GBM. Here, we will provide an overview of HATs, their dual roles and functions in cancer as both tumor suppressors and oncogenes and focus specifically on their implications in GBM resistance. We also discuss the technical challenges in developing selective HAT inhibitors and highlight their promise as potential anti-cancer therapeutics for treating intractable cancers such as GBM.


Subject(s)
Brain Neoplasms , Drug Resistance, Neoplasm , Glioblastoma , Histone Acetyltransferases , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/enzymology , Glioblastoma/pathology , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Epigenesis, Genetic , Molecular Targeted Therapy , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
4.
PLoS Negl Trop Dis ; 18(8): e0012428, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159234

ABSTRACT

BACKGROUND: Schistosomiasis is a relatively neglected parasitic disease that afflicts more than 250 million people worldwide, for which the control strategy relies mainly on mass treatment with the only available drug, praziquantel (PZQ). This approach is not sustainable and is a priority for developing novel drug candidates for the treatment and control of schistosomiasis. METHODOLOGYS/PRINCIPAL FINDINGS: In our previous study, we found that DW-3-15, a kind of PZQ derivative, could significantly downregulate the expression of the histone acetyltransferase of Schistosoma japonicum (SjHAT). In this study, several commercially available HAT inhibitors, A485, C646 and curcumin were screened in vitro to verify their antischistosomal activities against S. japonicum juveniles and adults. Parasitological studies and scanning electron microscopy were used to study the primary action characteristics of HAT inhibitors in vitro. Quantitative real-time PCR was employed to detect the mRNA level of SjHAT after treatment with different HAT inhibitors. Our results demonstrated that curcumin was the most effective inhibitor against both juveniles and adults of S. japonicum, and its schistosomicidal effects were time- and dose dependent. However, A485 and C646 had limited antischistosomal activity. Scanning electron microscopy demonstrated that in comparison with DW-3-15, curcumin caused similar tegumental changes in male adult worms. Furthermore, both curcumin and DW-3-15 significantly decreased the SjHAT mRNA level, and curcumin dose-dependently reduced the SjHAT expression level in female, male and juvenile worms. CONCLUSIONS: Among the three commercially available HATs, curcumin was the most potent against schistosomes. Both curcumin and our patent compound DW-3-15 markedly downregulated the expression of SjHAT, indicating that SjHAT may be a potential therapeutic target for developing novel antischistosomal drug candidates.


Subject(s)
Curcumin , Histone Acetyltransferases , Schistosoma japonicum , Animals , Schistosoma japonicum/drug effects , Curcumin/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Female , Male , Enzyme Inhibitors/pharmacology , Microscopy, Electron, Scanning , Real-Time Polymerase Chain Reaction , Mice , Schistosomicides/pharmacology
5.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 36(2): 207-214, 2024 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-38857968

ABSTRACT

Schistosomiasis is a neglected zoonotic parasitic disease. Currently, praziquantel is the drug of choice for the treatment of schistosomiasis, and is the only effective chemical for treatment of schistosomiasis japonica. Since its introduction in the 1970s, praziquantel has been used for large-scale chemotherapy of schistosomiasis for over 40 years. However, there have been reports pertaining to the resistance to praziquantel in schistosomes. Therefore, development of novel antischistosomal agents as alternatives of praziquantel, is of great need. Histone deacetylases and histone acetyltransferases have been recently reported to play critical roles in the growth, development and reproduction of schistosomes, and are considered as potential drug targets for the treatment of schistosomiasis. This review summarizes the latest advances of histone deacetylase and histone acetyltransferase inhibitors in the research on antischistosomal drugs, so as to provide insights into research and development of novelantischistosomal agents.


Subject(s)
Histone Acetyltransferases , Histone Deacetylase Inhibitors , Histone Deacetylases , Animals , Histone Deacetylase Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Humans , Histone Deacetylases/metabolism , Schistosoma/drug effects , Schistosoma/enzymology , Schistosoma/physiology , Schistosomiasis/drug therapy , Schistosomicides/pharmacology , Schistosomicides/therapeutic use
6.
J Agric Food Chem ; 72(27): 15176-15189, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38943677

ABSTRACT

Fusarium head blight caused by Fusarium graminearum is a devastating disease in wheat that seriously endangers food security and human health. Previous studies have found that the secondary metabolite phenazine-1-carboxamide produced by biocontrol bacteria inhibited F. graminearum by binding to and inhibiting the activity of histone acetyltransferase Gcn5 (FgGcn5). However, the detailed mechanism of this inhibition remains unknown. Our structural and biochemical studies revealed that phenazine-1-carboxamide (PCN) binds to the histone acetyltransferase (HAT) domain of FgGcn5 at its cosubstrate acetyl-CoA binding site, thus competitively inhibiting the histone acetylation function of the enzyme. Alanine substitution of the residues in the binding site shared by PCN and acetyl-CoA not only decreased the histone acetylation level of the enzyme but also dramatically impacted the development, mycotoxin synthesis, and virulence of the strain. Taken together, our study elucidated a competitive inhibition mechanism of Fusarium fungus by PCN and provided a structural template for designing more potent phenazine-based fungicides.


Subject(s)
Fungal Proteins , Fungicides, Industrial , Fusarium , Histone Acetyltransferases , Phenazines , Plant Diseases , Triticum , Fusarium/metabolism , Fusarium/drug effects , Fusarium/genetics , Phenazines/metabolism , Phenazines/pharmacology , Phenazines/chemistry , Fungal Proteins/metabolism , Fungal Proteins/genetics , Fungal Proteins/chemistry , Fungicides, Industrial/pharmacology , Fungicides, Industrial/chemistry , Fungicides, Industrial/metabolism , Plant Diseases/microbiology , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/antagonists & inhibitors , Triticum/microbiology , Binding Sites , Acetylation
7.
Nat Med ; 30(8): 2242-2250, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38824244

ABSTRACT

Inhibition of histone lysine acetyltransferases (KATs) KAT6A and KAT6B has shown antitumor activity in estrogen receptor-positive (ER+) breast cancer preclinical models. PF-07248144 is a selective catalytic inhibitor of KAT6A and KAT6B. In the present study, we report the safety, pharmacokinetics (PK), pharmacodynamics, efficacy and biomarker results from the first-in-human, phase 1 dose escalation and dose expansion study (n = 107) of PF-07248144 monotherapy and fulvestrant combination in heavily pretreated ER+ human epidermal growth factor receptor-negative (HER2-) metastatic breast cancer (mBC). The primary objectives of assessing the safety and tolerability and determining the recommended dose for expansion of PF-07248144, as monotherapy and in combination with fulvestrant, were met. Secondary endpoints included characterization of PK and evaluation of antitumor activity, including objective response rate (ORR) and progression-free survival (PFS). Common treatment-related adverse events (any grade; grades 3-4) included dysgeusia (83.2%, 0%), neutropenia (59.8%, 35.5%) and anemia (48.6%, 13.1%). Exposure was approximately dose proportional. Antitumor activity was observed as monotherapy. For the PF-07248144-fulvestrant combination (n = 43), the ORR (95% confidence interval (CI)) was 30.2% (95% CI = 17.2-46.1%) and the median PFS was 10.7 (5.3-not evaluable) months. PF-07248144 demonstrated a tolerable safety profile and durable antitumor activity in heavily pretreated ER+HER2- mBC. These findings establish KAT6A and KAT6B as druggable cancer targets, provide clinical proof of concept and reveal a potential avenue to treat mBC. clinicaltrial.gov registration: NCT04606446 .


Subject(s)
Breast Neoplasms , Fulvestrant , Histone Acetyltransferases , Receptor, ErbB-2 , Receptors, Estrogen , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Middle Aged , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Receptors, Estrogen/metabolism , Fulvestrant/therapeutic use , Fulvestrant/administration & dosage , Aged , Adult , Neoplasm Metastasis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
8.
Angew Chem Int Ed Engl ; 63(32): e202404645, 2024 08 05.
Article in English | MEDLINE | ID: mdl-38801173

ABSTRACT

Phenotypic assays detect small-molecule bioactivity at functionally relevant cellular sites, and inherently cover a variety of targets and mechanisms of action. They can uncover new small molecule-target pairs and may give rise to novel biological insights. By means of an osteoblast differentiation assay which employs a Hedgehog (Hh) signaling agonist as stimulus and which monitors an endogenous marker for osteoblasts, we identified a pyrrolo[3,4-g]quinoline (PQ) pseudo-natural product (PNP) class of osteogenesis inhibitors. The most potent PQ, termed Tafbromin, impairs canonical Hh signaling and modulates osteoblast differentiation through binding to the bromodomain 2 of the TATA-box binding protein-associated factor 1 (TAF1). Tafbromin is the most selective TAF1 bromodomain 2 ligand and promises to be an invaluable tool for the study of biological processes mediated by TAF1(2) bromodomains.


Subject(s)
TATA-Binding Protein Associated Factors , Transcription Factor TFIID , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/antagonists & inhibitors , Humans , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Biological Products/chemistry , Biological Products/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Cell Differentiation/drug effects , Quinolines/chemistry , Quinolines/pharmacology , Molecular Structure
9.
Expert Opin Ther Pat ; 34(4): 211-229, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38742308

ABSTRACT

INTRODUCTION: SMARCA2 and SMARCA4 are subunits of the SWI/SNF complex which is a chromatin remodeling complex and a key epigenetic regulator that facilitates gene expression. Tumors with loss of function mutations in SMARCA4 rely on SMARCA2 for cell survival and this synthetic lethality is a potential therapeutic strategy to treat cancer. AREAS COVERED: The current review focuses on patent applications that claim proteolysis-targeting chimeras (PROTAC) degraders that bind the bromodomain site of SMARCA2 and are published between January 2019-June 2023. A total of 29 applications from 9 different applicants were evaluated. EXPERT OPINION: SMARCA2/4 bromodomain inhibitors do not lead to desired effects on cancer proliferation; however, companies have converted bromodomain binders into PROTACs to degrade the protein, with a preference for SMARCA2 over SMARCA4. Selective degradation of SMARCA2 is most likely required to be efficacious in the SMARCA4-deficient setting, while allowing for sufficient safety margin in normal tissues. With several patent applications disclosed recently, interest in targeting SMARCA2 should continue, especially with a selective SMARCA2 PROTAC now in the clinic from Prelude Therapeutics. The outcome of the clinical trials will influence the evolution of selective SMARCA2 PROTACs development.


Subject(s)
Antineoplastic Agents , DNA Helicases , Neoplasms , Nuclear Proteins , Patents as Topic , Synthetic Lethal Mutations , Transcription Factors , Humans , Transcription Factors/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Animals , DNA Helicases/metabolism , Antineoplastic Agents/pharmacology , Proteolysis/drug effects , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Molecular Targeted Therapy
10.
Sci Rep ; 14(1): 11721, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38777823

ABSTRACT

It has recently been shown that KAT8, a genome-wide association study candidate risk gene for Parkinson's Disease, is involved in PINK1/Parkin-dependant mitophagy. The KAT8 gene encodes a lysine acetyltransferase and represents the catalytically active subunit of the non-specific lethal epigenetic remodelling complex. In the current study, we show that contrary to KAT5 inhibition, dual inhibition of KAT5 and KAT8 via the MG149 compound inhibits the initial steps of the PINK1-dependant mitophagy process. More specifically, our study shows that following mitochondrial depolarisation induced by mitochondrial toxins, MG149 treatment inhibits PINK1-dependant mitophagy initiation by impairing PINK1 activation, and subsequent phosphorylation of Parkin and ubiquitin. While this inhibitory effect of MG149 on PINK1-activation is potent, MG149 treatment in the absence of mitochondrial toxins is sufficient to depolarise the mitochondrial membrane, recruit PINK1 and promote partial downstream recruitment of the autophagy receptor p62, leading to an increase in mitochondrial delivery to the lysosomes. Altogether, our study provides additional support for KAT8 as a regulator of mitophagy and autophagy processes.


Subject(s)
Mitochondria , Mitophagy , Protein Kinases , Ubiquitin-Protein Ligases , Mitophagy/drug effects , Humans , Protein Kinases/metabolism , Protein Kinases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Phosphorylation/drug effects , Membrane Potential, Mitochondrial/drug effects , HeLa Cells
11.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557491

ABSTRACT

Mutations in genes encoding chromatin modifiers are enriched among mutations causing intellectual disability. The continuing development of the brain postnatally, coupled with the inherent reversibility of chromatin modifications, may afford an opportunity for therapeutic intervention following a genetic diagnosis. Development of treatments requires an understanding of protein function and models of the disease. Here, we provide a mouse model of Say-Barber-Biesecker-Young-Simpson syndrome (SBBYSS) (OMIM 603736) and demonstrate proof-of-principle efficacy of postnatal treatment. SBBYSS results from heterozygous mutations in the KAT6B (MYST4/MORF/QFK) gene and is characterized by intellectual disability and autism-like behaviors. Using human cells carrying SBBYSS-specific KAT6B mutations and Kat6b heterozygous mice (Kat6b+/-), we showed that KAT6B deficiency caused a reduction in histone H3 lysine 9 acetylation. Kat6b+/- mice displayed learning, memory, and social deficits, mirroring SBBYSS individuals. Treatment with a histone deacetylase inhibitor, valproic acid, or an acetyl donor, acetyl-carnitine (ALCAR), elevated histone acetylation levels in the human cells with SBBYSS mutations and in brain and blood cells of Kat6b+/- mice and partially reversed gene expression changes in Kat6b+/- cortical neurons. Both compounds improved sociability in Kat6b+/- mice, and ALCAR treatment restored learning and memory. These data suggest that a subset of SBBYSS individuals may benefit from postnatal therapeutic interventions.


Subject(s)
Abnormalities, Multiple , Acetylcarnitine , Congenital Hypothyroidism , Craniofacial Abnormalities , Histone Acetyltransferases , Intellectual Disability , Joint Instability , Animals , Humans , Mice , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/genetics , Acetylation , Acetylcarnitine/pharmacology , Acetylcarnitine/therapeutic use , Blepharophimosis , Chromatin , Craniofacial Abnormalities/drug therapy , Craniofacial Abnormalities/genetics , Exons , Facies , Heart Defects, Congenital , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Intellectual Disability/drug therapy , Intellectual Disability/genetics
12.
J Transl Med ; 21(1): 31, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36650543

ABSTRACT

NOC2 like nucleolar associated transcriptional repressor (NOC2L) was recently identified as a novel inhibitor of histone acetyltransferase (INHAT). NOC2L is found to have two INHAT function domains and regulates histone acetylation in a histone deacetylases (HDAC) independent manner, which is distinct from other INHATs. In this review, we summarize the biological function of NOC2L in histone acetylation regulation, P53-mediated transcription, ribosome RNA processing, certain development events and carcinogenesis. We propose that NOC2L may be explored as a potential biomarker and a therapeutic target in clinical practice.


Subject(s)
Histone Acetyltransferases , Histones , Repressor Proteins , Acetylation , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histones/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics
13.
J Am Chem Soc ; 144(2): 832-844, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34985906

ABSTRACT

Owing to its roles in human health and disease, the modification of nuclear, cytoplasmic, and mitochondrial proteins with O-linked N-acetylglucosamine residues (O-GlcNAc) has emerged as a topic of great interest. Despite the presence of O-GlcNAc on hundreds of proteins within cells, only two enzymes regulate this modification. One of these enzymes is O-GlcNAcase (OGA), a dimeric glycoside hydrolase that has a deep active site cleft in which diverse substrates are accommodated. Chemical tools to control OGA are emerging as essential resources for helping to decode the biochemical and cellular functions of the O-GlcNAc pathway. Here we describe rationally designed bicyclic thiazolidine inhibitors that exhibit superb selectivity and picomolar inhibition of human OGA. Structures of these inhibitors in complex with human OGA reveal the basis for their exceptional potency and show that they extend out of the enzyme active site cleft. Leveraging this structure, we create a high affinity chemoproteomic probe that enables simple one-step purification of endogenous OGA from brain and targeted proteomic mapping of its post-translational modifications. These data uncover a range of new modifications, including some that are less-known, such as O-ubiquitination and N-formylation. We expect that these inhibitors and chemoproteomics probes will prove useful as fundamental tools to decipher the mechanisms by which OGA is regulated and directed to its diverse cellular substrates. Moreover, the inhibitors and structures described here lay out a blueprint that will enable the creation of chemical probes and tools to interrogate OGA and other carbohydrate active enzymes.


Subject(s)
Antigens, Neoplasm/metabolism , Bridged Bicyclo Compounds/chemistry , Enzyme Inhibitors/chemistry , Histone Acetyltransferases/metabolism , Hyaluronoglucosaminidase/metabolism , Amino Acid Sequence , Brain/metabolism , Bridged Bicyclo Compounds/metabolism , Catalytic Domain , Chromatography, High Pressure Liquid , Enzyme Inhibitors/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Mass Spectrometry , Peptides/analysis , Peptides/chemistry , Protein Processing, Post-Translational , Proteomics/methods , Structure-Activity Relationship , Thiazolidines/chemistry , Thiazolidines/metabolism , beta-Hexosaminidase alpha Chain/antagonists & inhibitors , beta-Hexosaminidase alpha Chain/metabolism
14.
PLoS One ; 16(12): e0261388, 2021.
Article in English | MEDLINE | ID: mdl-34914791

ABSTRACT

Cardiac hypertrophy is a complex process induced by the activation of multiple signaling pathways. We previously reported that anacardic acid (AA), a histone acetyltransferase (HAT) inhibitor, attenuates phenylephrine (PE)-induced cardiac hypertrophy by downregulating histone H3 acetylation at lysine 9 (H3K9ac). Unfortunately, the related upstream signaling events remained unknown. The mitogen-activated protein kinase (MAPK) pathway is an important regulator of cardiac hypertrophy. In this study, we explored the role of JNK/MAPK signaling pathway in cardiac hypertrophy induced by PE. The mice cardiomyocyte hypertrophy model was successfully established by treating cells with PE in vitro. This study showed that p-JNK directly interacts with HATs (P300 and P300/CBP-associated factor, PCAF) and alters H3K9ac. In addition, both the JNK inhibitor SP600125 and the HAT inhibitor AA attenuated p-JNK overexpression and H3K9ac hyperacetylation by inhibiting P300 and PCAF during PE-induced cardiomyocyte hypertrophy. Moreover, we demonstrated that both SP600125 and AA attenuate the overexpression of cardiac hypertrophy-related genes (MEF2A, ANP, BNP, and ß-MHC), preventing cardiomyocyte hypertrophy and dysfunction. These results revealed a novel mechanism through which AA might protect mice from PE-induced cardiomyocyte hypertrophy. In particular, AA inhibits the effects of JNK signaling on HATs-mediated histone acetylation, and could therefore be used to prevent and treat pathological cardiac hypertrophy.


Subject(s)
Anacardic Acids/pharmacology , Cardiomegaly/physiopathology , MAP Kinase Signaling System/physiology , Acetylation , Anacardic Acids/metabolism , Animals , Anthracenes/pharmacology , Cardiomegaly/metabolism , China , Disease Models, Animal , Female , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histones/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenylephrine/adverse effects , Phenylephrine/pharmacology , Primary Cell Culture , Signal Transduction/drug effects , p300-CBP Transcription Factors
15.
Pharmacol Res ; 174: 105930, 2021 12.
Article in English | MEDLINE | ID: mdl-34626770

ABSTRACT

Hematological malignancies, unlike solid tumors, are a group of malignancies caused by abnormal differentiation of hematopoietic stem cells. Monocytic leukemia zinc finger protein (MOZ), a member of the MYST (MOZ, Ybf2/Sas3, Sas2, Tip60) family, is a histone acetyltransferase. MOZ is involved in various cellular functions: generation and maintenance of hematopoietic stem cells, development of erythroid cells, B-lineage progenitors and myeloid cells, and regulation of cellular senescence. Studies have shown that MOZ is susceptible to translocation in chromosomal rearrangements to form fusion genes, leading to the fusion of MOZ with other cellular regulators to form MOZ fusion proteins. Different MOZ fusion proteins have different roles, such as in the development and progression of hematological malignancies and inhibition of cellular senescence. Thus, MOZ is an attractive target, and targeting MOZ to design small-molecule drugs can help to treat hematological malignancies. This review summarizes recent progress in biology and medicinal chemistry for the histone acetyltransferase MOZ. In the biology section, MOZ and cofactors, structures of MOZ and related HATs, MOZ and fusion proteins, and roles of MOZ in cancer are discussed. In medicinal chemistry, recent developments in MOZ inhibitors are summarized.


Subject(s)
Hematologic Neoplasms , Histone Acetyltransferases , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans
17.
Mol Cell Endocrinol ; 537: 111425, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34391847

ABSTRACT

BACKGROUND: Previously, we reported that Mof was highly expressed in α-cells, and its knockdown led to ameliorated fasting blood glucose (FBG) and glucose tolerance in non-diabetic mice, attributed by reduced total α-cell but enhanced prohormone convertase (PC)1/3-positive α-cell mass. However, how Mof and histone 4 lysine 16 acetylation (H4K16ac) control α-cell and whether Mof inhibition improves glucose handling in type 2 diabetes (T2DM) mice remain unknown. METHODS: Mof overexpression and chromatin immunoprecipitation sequence (ChIP-seq) based on H4K16ac were applied to determine the effect of Mof on α-cell transcriptional factors and underlying mechanism. Then we administrated mg149 to α-TC1-6 cell line, wild type, db/db and diet-induced obesity (DIO) mice to observe the impact of Mof inhibition in vitro and in vivo. In vitro, western blotting and TUNEL staining were used to examine α-cell apoptosis and function. In vivo, glucose tolerance, hormone levels, islet population, α-cell ratio and the co-staining of glucagon and PC1/3 or PC2 were examined. RESULTS: Mof activated α-cell-specific transcriptional network. ChIP-seq results indicated that H4K16ac targeted essential genes regulating α-cell differentiation and function. Mof activity inhibition in vitro caused impaired α-cell function and enhanced apoptosis. In vivo, it contributed to ameliorated glucose intolerance and islet dysfunction, characterized by decreased fasting glucagon and elevated post-challenge insulin levels in T2DM mice. CONCLUSION: Mof regulates α-cell differentiation and function via acetylating H4K16ac and H4K16ac binding to Pax6 and Foxa2 promoters. Mof inhibition may be a potential interventional target for T2DM, which led to decreased α-cell ratio but increased PC1/3-positive α-cells.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Secreting Cells/enzymology , Glucagon-Secreting Cells/pathology , Glucose Intolerance/enzymology , Glucose Intolerance/physiopathology , Histone Acetyltransferases/antagonists & inhibitors , Acetylation/drug effects , Animals , Apoptosis/drug effects , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diet , Gene Regulatory Networks/drug effects , Glucagon-Secreting Cells/drug effects , Histone Acetyltransferases/metabolism , Histones/metabolism , Lysine/metabolism , Mice, Inbred C57BL , Obesity/etiology , Proprotein Convertase 1/metabolism , Salicylates/pharmacology
18.
J Pharmacol Sci ; 147(2): 169-175, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34384564

ABSTRACT

INTRODUCTION: Heart failure is the final pathway for a wide spectrum of myocardial stress, including hypertension and myocardial infarction. However, the potential effects of metformin on cardiac hypertrophy are still unclear. PURPOSE: The purpose of this study was to investigate whether metformin leads to suppression of hypertrophic responses in cardiomyocytes. METHODS AND RESULTS: To investigate whether metformin inhibited p300-histone acetyltransferase (HAT), we performed an in vitro HAT assay. Metformin directly inhibited p300-mediated acetylation of histone-H3K9. To examine the effects of metformin on hypertrophic responses, cardiomyocytes prepared from neonatal rats were treated with metformin and stimulated with saline or phenylephrine (PE), a α1-adrenergic agonist for 48 h. PE stimulus showed an increase in cell size, myofibrillar organization, expression of the endogenous atrial natriuretic factor and brain natriuretic peptide genes, and acetylation of histone-H3K9 compared with saline-treated cells. These PE-induced changes were inhibited by metformin. Next, to examine the effect of metformin on p300-mediated hypertrophy, cardiomyocytes were transfected with expression vector of p300. Metformin significantly suppressed p300-induced hypertrophic responses and acetylation of histone-H3K9. CONCLUSIONS: The study demonstrates that metformin can suppress PE-induced and p300-mediated hypertrophic responses. Metformin may be useful for the treatment of patients with diabetes and heart failure.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/adverse effects , Cardiomegaly/chemically induced , Cardiomegaly/pathology , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Metformin/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenylephrine/adverse effects , Acetylation/drug effects , Animals , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cells, Cultured , Heart Failure/drug therapy , Heart Failure/etiology , Metformin/therapeutic use , Rats, Sprague-Dawley
19.
Clin Transl Med ; 11(7): e495, 2021 07.
Article in English | MEDLINE | ID: mdl-34323404

ABSTRACT

Castration-resistant prostate cancer (CRPC) is the latest stage of PCa, and there is almost no effective treatment available for the patients with CRPC when next-generation androgen deprivation therapy drugs, such as enzalutamide (ENZ), fail. The androgen receptor (AR) plays key roles in PCa and CRPC progression and drug resistance. Histone acetyltransferase 1 (HAT1) has recently been reported to be highly expressed in some tumors, such as lung carcinoma. However, what relationship between the AR and HAT1, and whether or how HAT1 plays roles in CRPC progression and drug resistance remain elusive. In the present study, we found that HAT1 is highly expressed in PCa cells, and the overexpression of HAT1 is linked with CRPC cell proliferation. Moreover, the HAT1 expression is positively correlated with the expression of AR, including both AR-FL (full-length) and AR-V7 (variant 7), which is mainly mediated by a bromodomain containing protein 4 (BRD4) -mediated pathway. Furthermore, knockdown of HAT1 can re-sensitize the response of CRPC cells to ENZ treatment in cells and mouse models. In addition, ascorbate was observed to decrease AR expression through downregulation of HAT1 expression. Collectively, our findings reveal a novel AR signaling regulation pathway in PCa and CRPC and suggest that HAT1 serves as a critical oncoprotein and an ideal target for the treatment of ENZ resistance in CRPC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Histone Acetyltransferases/metabolism , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Humans , Male , Mice , Mice, Inbred NOD , Nitriles/therapeutic use , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Receptors, Androgen/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
20.
Eur J Med Chem ; 223: 113649, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34186233

ABSTRACT

O-GlcNAcylation is a dynamic post-translational modification mediated by O-linked ß-N-acetylglucosamine transferase (OGT) and O-GlcNAc hydrolase (OGA), that adds or removes a single ß-N-acetylglucosamine (GlcNAc) moiety to or from serine/threonine residues of nucleocytosolic and mitochondrial proteins, respectively. The perturbed homeostasis of O-GlcNAc cycling results in several pathological conditions. Human OGA is a promising therapeutic target in diseases where aberrantly low levels of O-GlcNAc are experienced, such as tauopathy in Alzheimer's disease. A new class of potent OGA inhibitors, 2-acetamido-2-deoxy-d-glucono-1,5-lactone (thio)semicarbazones, have been identified. Eight inhibitors were designed and synthesized in five steps starting from d-glucosamine and with 15-55% overall yields. A heterologous OGA expression protocol with strain selection and isolation has been optimized that resulted in stable, active and full length human OGA (hOGA) isomorph. Thermal denaturation kinetics of hOGA revealed environmental factors affecting hOGA stability. From kinetics experiments, the synthesized compounds proved to be efficient competitive inhibitors of hOGA with Ki-s in the range of ∼30-250 nM and moderate selectivity with respect to lysosomal ß-hexosaminidases. In silico studies consisting of Prime protein-ligand refinements, QM/MM optimizations and QM/MM-PBSA binding free energy calculations revealed the factors governing the observed potencies, and led to design of the most potent analogue 2-acetamido-2-deoxy-d-glucono-1,5-lactone 4-(2-naphthyl)-semicarbazone 6g (Ki = 36 nM). The protocol employed has applications in future structure based inhibitor design targeting OGA.


Subject(s)
Antigens, Neoplasm/metabolism , Enzyme Inhibitors/chemistry , Histone Acetyltransferases/metabolism , Hyaluronoglucosaminidase/metabolism , Lactones/chemistry , Semicarbazones/chemistry , Antigens, Neoplasm/genetics , Binding Sites , Enzyme Inhibitors/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/genetics , Kinetics , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Stability , Quantum Theory , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Semicarbazones/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL