Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 240
Filter
1.
J Nat Prod ; 87(4): 1187-1196, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38632902

ABSTRACT

Psammaplins are sulfur containing bromotyrosine alkaloids that have shown antitumor activity through the inhibition of class I histone deacetylases (HDACs). The cytotoxic properties of psammaplin A (1), the parent compound, are related to peroxisome proliferator-activated receptor γ (PPARγ) activation, but the mechanism of action of its analogs psammaplin K (2) and bisaprasin (3) has not been elucidated. In this study, the protective effects against oxidative stress of compounds 1-3, isolated from the sponge Aplysinella rhax, were evaluated in SH-SY5Y cells. The compounds improved cell survival, recovered glutathione (GSH) content, and reduced reactive oxygen species (ROS) release at nanomolar concentrations. Psammaplins restored mitochondrial membrane potential by blocking mitochondrial permeability transition pore opening and reducing cyclophilin D expression. This effect was mediated by the capacity of 1-3 to activate PPARγ, enhancing gene expression of the antioxidant enzymes catalase, nuclear factor E2-related factor 2 (Nrf2), and glutathione peroxidase. Finally, HDAC3 activity was reduced by 1-3 under oxidative stress conditions. This work is the first description of the neuroprotective activity of 1 at low concentrations and the mechanism of action of 2 and 3. Moreover, it links for the first time the previously described effects of 1 in HDAC3 and PPARγ signaling, opening a new research field for the therapeutic potential of this compound family.


Subject(s)
Disulfides , Oxidative Stress , PPAR gamma , Tyrosine/analogs & derivatives , PPAR gamma/metabolism , Oxidative Stress/drug effects , Humans , Animals , Molecular Structure , Reactive Oxygen Species/metabolism , Neurons/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/drug effects , NF-E2-Related Factor 2/metabolism , Porifera/chemistry , Membrane Potential, Mitochondrial/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Glutathione/metabolism , Alkaloids/pharmacology , Alkaloids/chemistry , Catalase/metabolism , Glutathione Peroxidase/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism
2.
J Pharm Pharm Sci ; 25: 24-40, 2022.
Article in English | MEDLINE | ID: mdl-34995473

ABSTRACT

With the significant increase of patients suffering from different types of cancer, it is evident that prompt measures in the development of novel and effective agents need to be taken. Pyrrole moiety has been found in various active compounds with anti-inflammatory, antiseptic, antibacterial, lipid-lowering and anticancer properties. Recent advances in the exploration of highly active and selective cytotoxic structures containing pyrrole motifs have shown promising data for future investigations. Accordingly, this review presents an overview of recent developments in the pyrrole derivatives as anticancer agents, with a main focus towards the key moieties required for the anti-tumor activities. Pyrrole molecules comprising prominent targeting capacities against microtubule polymerization, tyrosine kinases, cytochrome p450 family 1, histone deacetylase and bcl-2 proteins were reported. In addition, several mechanisms of action, such as apoptosis, cell cycle arrest, inhibiting kinases, angiogenesis, disruption of cell migration, modulation of nuclear receptor responsiveness and others were analyzed. Furthermore, in most of the discussed cases we provided synthesis schemes of the mentioned molecules. Overall, the utilization of pyrrole scaffold for the design and synthesis of novel anticancer drugs could be a promising approach for future investigations.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Pyrroles/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cytochrome P-450 Enzyme System/drug effects , Genes, bcl-2/drug effects , Histone Deacetylases/drug effects , Humans , Microtubules/drug effects , Protein-Tyrosine Kinases/drug effects , Pyrroles/chemistry , Structure-Activity Relationship
3.
Neuropharmacology ; 204: 108893, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34822816

ABSTRACT

Microduplication of the human 16p11.2 gene locus is associated with a range of neurodevelopmental outcomes, including autism spectrum disorder (ASD). Mice carrying heterozygous 16p11.2 duplication (16p11.2dp/+) display social deficits, which is attributable to impaired GABAergic synaptic function in prefrontal cortex (PFC) driven by downregulation of Npas4, an activity-dependent transcription factor that regulates GABA synapse formation. However, the molecular mechanisms underlying the diminished transcription of Npas4 in 16p11.2 duplication remain unknown. Npas4 is one of the target genes regulated by histone deacetylase 5 (HDAC5), an epigenetic enzyme repressing gene expression via removal of transcription-permissive acetyl groups from histones. Here we report that HDAC5 expression is elevated and histone acetylation is reduced at the Npas4 promoter in PFC of 16p11.2dp/+ mice. Treatment with the HDAC5 inhibitor LMK235 normalizes histone acetylation, restores GABAergic signaling in PFC, and significantly improves social preference in 16p11.2dp/+ mice. These findings suggest that HDAC5 inhibition is a promising therapeutic avenue to alleviate genetic, synaptic and behavioral deficits in 16p11.2 duplication conditions.


Subject(s)
Autistic Disorder/drug therapy , Autistic Disorder/genetics , Benzamides/pharmacology , Benzamides/therapeutic use , Chromosome Disorders/drug therapy , Chromosome Disorders/genetics , Histone Deacetylases/drug effects , Histone Deacetylases/physiology , Intellectual Disability/drug therapy , Intellectual Disability/genetics , Acetylation/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Disease Models, Animal , Down-Regulation , Gene Expression , Gene Expression Regulation, Enzymologic , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice, Transgenic
4.
Clin Sci (Lond) ; 136(1): 45-60, 2022 01 14.
Article in English | MEDLINE | ID: mdl-34918039

ABSTRACT

Cold storage/rewarming is an inevitable process for kidney transplantation from deceased donors, which correlates closely with renal ischemia-reperfusion injury (IRI) and the occurrence of delayed graft function. Histone deacetylases (HDAC) are important epigenetic regulators, but their involvement in cold storage/rewarming injury in kidney transplantation is unclear. In the present study, we showed a dynamic change of HDAC3 in a mouse model of kidney cold storage followed by transplantation. We then demonstrated that the selective HDAC3 inhibitor RGFP966 could reduce acute tubular injury and cell death after prolonged cold storage with transplantation. RGFP966 also improved renal function, kidney repair and tubular integrity when the transplanted kidney became the sole life-supporting graft in the recipient mouse. In vitro, cold storage of proximal tubular cells followed by rewarming induced remarkable cell death, which was suppressed by RGFP966 or knockdown of HDAC3 with shRNA. Inhibition of HDAC3 decreased the mitochondrial pathway of apoptosis and preserved mitochondrial membrane potential. Collectively, HDAC3 plays a pathogenic role in cold storage/rewarming injury in kidney transplantation, and its inhibition may be a therapeutic option.


Subject(s)
Acrylamides/therapeutic use , Histone Deacetylases/drug effects , Kidney Transplantation , Phenylenediamines/therapeutic use , Reperfusion Injury/prevention & control , Allografts , Animals , Apoptosis , Cold Temperature , Gene Knockdown Techniques , Histone Deacetylases/genetics , Kidney Tubules, Proximal/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Organ Preservation/adverse effects , RNA, Small Interfering
5.
Int J Immunopathol Pharmacol ; 35: 20587384211034086, 2021.
Article in English | MEDLINE | ID: mdl-34344210

ABSTRACT

INTRODUCTION: Cruciferous vegetables are a rich source of sulforaphane (SFN), which acts as a natural HDAC inhibitor (HDACi). Our previous study found that HDACi could restore histone acetyltransferase/histone deacetylase (HAT/HDAC) balance in the cochlea and attenuate gentamicin-induced hearing loss in guinea pigs. Here, we investigated the protective effect of SFN on cisplatin-induced hearing loss (CIHL). METHODS: Thirty rats were randomly divided into 3 equal groups: the control group, cisplatin group, and SFN+cisplatin group. Rats were injected with SFN (30 mg/kg once a day) and cisplatin (7 mg/kg twice a day) for 7 days to investigate the protective role of SFN on CIHL. We observed auditory brainstem response (ABR) threshold shifts and immunostained cochlear basilar membranes of rats. For in vitro experiments, we treated HEI-OC1 cells and rat cochlear organotypic cultures with SFN (5, 10, and 15 µM) and cisplatin (10 µM). Immunofluorescence, cell viability, and protein analysis were performed to further analyze the protective mechanism of SFN on CIHL. RESULTS: SFN (30 mg/kg once a day) decreased cisplatin (7 mg/kg twice a day)-induced ABR threshold shifts and outer hair cell loss. CCK-8 assay showed that cisplatin (10 µM) reduced the viability of HEI-OC1 cells to 42%, and SFN had a dose-dependent protective effect. In cochlear organotypic cultures, we found that SFN (10 and 15 µM) increased cisplatin (10 µM)-induced myosin 7a+ cell count and restored ciliary morphology. SFN (5, 10, and 15 µM) reversed the cisplatin (10 µM)-induced increase in HDAC2, -4, and -5 and SFN (15 µM) reversed the cisplatin (10 µM)-induced decrease in H3-Ack9 [acetyl-histone H3 (Lys9)] protein expression in HEI-OC1 cells. Neither cisplatin nor cisplatin combined with SFN affected the expression of HDAC7, or HDAC9. CONCLUSION: SFN prevented disruption of the HAT/HDAC balance, protecting against CIHL in rats.


Subject(s)
Antineoplastic Agents , Cisplatin , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Isothiocyanates/therapeutic use , Sulfoxides/therapeutic use , Animals , Cell Count , Cilia/pathology , Cochlea/pathology , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Outer/pathology , Histone Deacetylases/biosynthesis , Histone Deacetylases/drug effects , Histone Deacetylases/genetics , Rats , Rats, Wistar
6.
Biomed Pharmacother ; 142: 112021, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34463268

ABSTRACT

Valproic acid (VPA) is an approved drug for managing epileptic seizures, bipolar disorders, and migraine. VPA has been shown to elevate the level of gamma-aminobutyric acid (GABA) in the brain through competitive inhibition of GABA transaminase, thus promoting the availability of synaptic GABA and facilitating GABA-mediated responses. VPA, which is a small chain of fatty acids, prevents histone deacetylases (HDACs). HDACs play a crucial role in chromatin remodeling and gene expression through posttranslational changes of chromatin-associated histones. Recent studies reported a possible effect of VPA against particular types of cancers. This effect was partially attributed to its role in regulating epigenetic modifications through the inhibition of HDACs, which affect the expression of genes associated with cell cycle control, cellular differentiation, and apoptosis. In this review, we summarize the current information on the actions of VPA in diseases such as diabetes mellitus, kidney disorders, neurodegenerative diseases, muscular dystrophy, and cardiovascular disorders.


Subject(s)
GABA Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Valproic Acid/pharmacology , Animals , Anticonvulsants/pharmacology , Apoptosis/drug effects , Epigenesis, Genetic , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans
7.
Clin Sci (Lond) ; 135(14): 1751-1765, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34282832

ABSTRACT

Epigenetic dysregulation has long been identified as a key driver of leukemogenesis in acute myeloid leukemia (AML). However, epigenetic drugs such as histone deacetylase inhibitors (HDACis) targeting epigenetic alterations in AML have obtained only limited clinical efficiency without clear mechanism. Fortunately, we screened out a novel epigenetic agent named Apigenin-Vorinostat-Conjugate (AVC), which provides us a possibility to handle the heterogeneous malignancy. Its inhibition on HDACs was presented by HDACs expression, enzyme activity, and histone acetylation level. Its efficacy against AML was detected by cell viability assay and tumor progression of AML mouse model. Apoptosis is the major way causing cell death. We found that AVC efficiently suppresses leukemogenesis while sparing the normal human cells. Kasumi-1 cells are at least 20-fold higher sensitive to AVC (IC50 = 0.024 µM) than vorinostat (IC50 = 0.513 µM) and Ara-C (IC50 = 0.4366 µM). Furthermore, it can efficiently regress the tumorigenesis in AML mouse model while keeping the pivotal organs safe, demonstrating a feasibility and favorable safety profile in treatment of AML. Collectively, these preclinical data suggest a promising potential utilizing flavonoid-HDACi-conjugate as a next-generation epigenetic drug for clinical therapy against AML.


Subject(s)
Epigenesis, Genetic/drug effects , Flavonoids/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice
8.
Behav Brain Res ; 414: 113477, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34302880

ABSTRACT

Recently, epigenetic mechanisms are considered as the new potential targets for addiction treatment. This research was designed to explore the effect of histone acetylation on ΔFosB gene expression in morphine-induced conditioned place preference (CPP) in male rats. CPP was induced via morphine injection (5 mg/kg) for three consecutive days. Animals received low-dose theophylline (LDT) or Suberoylanilide Hydroxamic acid (SAHA), as an histone deacetylase (HDAC) activator or inhibitor, respectively, and a combination of both in subsequent extinction days. Following extinction, a priming dose of morphine (1 mg/kg) was administered to induce reinstatement. H4 acetylation and ΔFosB expression in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were assessed on the last day of extinction and the following CPP reinstatement. Our results demonstrated that daily administration of SAHA (25 mg/kg; i.p.), facilitated morphine-extinction and decreased CPP score in reinstatement of place preference. Conversely, injections of LDT (20 mg/kg; i.p.) prolonged extinction in animals. Co-administration of LDT and SAHA on extinction days counterbalanced each other, such that maintenance and reinstatement were no different than the control group. The gene expression of ΔFosB was increased by SAHA in NAc and mPFC compared to the control group. Administration of SAHA during extinction days, also altered histone acetylation in the NAc and mPFC on the last day of extinction, but not on reinstatement day. Collectively, administration of SAHA facilitated extinction and reduced reinstatement of morphine-induced CPP in rats. This study confirms the essential role of epigenetic mechanisms, specifically histone acetylation, in regulating drug-induced plasticity and seeking behaviors.


Subject(s)
Behavior, Animal , Conditioning, Classical , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Histones/metabolism , Morphine/pharmacology , Narcotics/pharmacology , Nucleus Accumbens , Prefrontal Cortex , Proto-Oncogene Proteins c-fos , Theophylline/pharmacology , Acetylation , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Histone Deacetylase Inhibitors/administration & dosage , Male , Morphine/administration & dosage , Narcotics/administration & dosage , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Theophylline/administration & dosage , Vorinostat/pharmacology
9.
Toxicology ; 458: 152830, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34097993

ABSTRACT

Previous studies have suggested that bisphenol A (BPA) has a toxic effect on bone development; however, its pathological mechanism has not been fully elucidated. In the present study, pregnant Wistar rats were intragastrically administered BPA (10 µg/kg per day) during gestational days 14-21. Then, bone tissues were obtained from neonatal rats on postnatal day 1 for histological analysis, and the bone mass of adult rat offspring was analyzed by micro-CT at postnatal week 10. Furthermore, osteoprogenitors from neonatal rats were obtained and treated with various concentrations of BPA in vitro to clarify the associated mechanism. In vivo, we found that prenatal BPA exposure reduced body weight and body length in female neonatal rats but not in male neonatal rats. Meanwhile, BPA exposure during pregnancy delayed bone development and reduced bone mass only in female rat offspring. Moreover, BPA exposure during pregnancy inhibited osteogenic function and downregulated the transforming growth factor ß (TGF ß) signaling pathway in the bone tissue of female neonatal rats. Our in vitro findings further indicated that various concentrations of BPA suppressed the osteogenic function of osteoprogenitors by downregulating the TGFß signaling pathway. Meanwhile, BPA downregulated H3K9ac and expression levels of TGFß via the ERß/HDAC5 signaling pathway. Collectively, this research revealed that prenatal BPA exposure impairs bone development and bone mass accumulation in female rat offspring, which was attributed to inhibitory osteogenic function via the ERß/HDAC5/TGFß signaling pathway.


Subject(s)
Benzhydryl Compounds/toxicity , Bone Development/drug effects , Bone and Bones/anatomy & histology , Endocrine Disruptors/toxicity , Estrogen Receptor beta/drug effects , Histone Deacetylases/drug effects , Phenols/toxicity , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Animals , Animals, Newborn , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Down-Regulation/drug effects , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Stem Cells/drug effects , X-Ray Microtomography
10.
Drug Discov Today ; 26(10): 2303-2314, 2021 10.
Article in English | MEDLINE | ID: mdl-33895313

ABSTRACT

Gastrointestinal (GI) malignancies account for substantial mortality and morbidity worldwide. They are generally promoted by dysregulated signal transduction and epigenetic pathways, which are controlled by specific enzymes. Recent studies demonstrated that histone deacetylases (HDACs) together with DNA methyltransferases (DNMTs) have crucial roles in the signal transduction/epigenetic pathways in GI regulation. In this review, we discuss various enzyme targets and their functional mechanisms responsible for the regulatory processes of GI malignancies. We also discuss the epigenetic therapeutic targets that are mainly facilitated by DNMT and HDAC inhibitors, which have functional consequences and clinical outcomes for GI malignancies.


Subject(s)
Epigenesis, Genetic , Gastrointestinal Neoplasms/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , DNA Modification Methylases/antagonists & inhibitors , Gastrointestinal Neoplasms/enzymology , Gastrointestinal Neoplasms/genetics , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Molecular Targeted Therapy
11.
Neuroreport ; 32(6): 423-430, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33788811

ABSTRACT

Behavioral sensitization, an animal model of drug addiction, persists for a prolonged period after repeated exposure to drugs of abuse. The persistence of an addiction behavioral phenotype suggests long-lasting changes in gene regulation at the epigenetic level. We measured the expression of histone deacetylases (HDACs) isoforms in the prefrontal cortex and dorsal striatum following the development of sensitization to cocaine (15 mg/kg, administered five times) and ethanol (0.5 g/kg, administered 15 times) to investigate the epigenetic changes that mediate sensitization. Animals sensitized to ethanol exhibited augmented locomotor activity in response to the cocaine challenge. Similarly, those sensitized to cocaine exhibited increased locomotor activity in response to an ethanol challenge. These findings indicate cross-sensitization between ethanol and cocaine and suggest that a common molecular mechanism underlying the cross-sensitization. In animals sensitized to cocaine or ethanol, mRNA levels of class II HDACs (HDAC4 and HDAC5) were decreased in the prefrontal cortex and dorsal striatum, whereas acute treatments with either drug had no effect on the expression of class II HDACs. By contrast, class I HDACs (HDAC1 and HDAC2) responded to the acute cocaine challenge, whereas sensitization itself did not have a consistent effect on class I HDAC levels. These findings support the hypothesis of a common epigenetic mechanism underlying persistent behavioral sensitization induced by different drugs, which may be mediated by the altered expression of class II HDACs.


Subject(s)
Brain/drug effects , Central Nervous System Depressants/pharmacology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Ethanol/pharmacology , Histone Deacetylases/drug effects , RNA, Messenger/drug effects , Alcoholism/genetics , Alcoholism/metabolism , Animals , Brain/metabolism , Central Nervous System Sensitization/drug effects , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Disease Models, Animal , Epigenesis, Genetic , Histone Deacetylase 1/drug effects , Histone Deacetylase 1/genetics , Histone Deacetylase 2/drug effects , Histone Deacetylase 2/genetics , Histone Deacetylases/genetics , Male , Neostriatum/drug effects , Neostriatum/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Transcriptome
12.
Oncogene ; 40(10): 1836-1850, 2021 03.
Article in English | MEDLINE | ID: mdl-33564072

ABSTRACT

Despite the widespread use of the blockade of immune checkpoints, for a significant number of cancer patients, these therapies have proven ineffective, presumably due to the immunosuppressive nature of the tumor microenvironment (TME). Critical drivers of immune escape in the TME include tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which not only mediate immune suppression, but also facilitate metastatic dissemination and impart resistance to immunotherapies. Thus, strategies that convert them into tumor fighters may offer great therapeutic potential. In this study, we evaluated whether pharmacologic modulation of macrophage phenotype by HDAC inhibitors (HDACi) could produce an anti-tumor effect. We demonstrated that low-dose HDACi trichostatin-A (TSA) markedly reshaped the tumor immune microenvironment by modulating the suppressive activity of infiltrating macrophages and inhibiting the recruitment of MDSCs in various tumors. These actions, in turn, augmented anti-tumor immune responses and further enhanced anti-tumor effects of immunotherapies. HDAC inhibition, however, also upregulated PD-L1, thereby limiting the beneficial therapeutic effects. Indeed, combining low-dose TSA with anti-PD-L1 in this model significantly enhanced the durability of tumor reduction and prolonged survival of tumor-bearing mice, compared with the effect of either treatment alone. These data introduce HDAC inhibition as a potential means to harness the anti-tumor potential of macrophages in cancer therapy.


Subject(s)
B7-H1 Antigen/genetics , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Melanoma, Experimental/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , Disease Models, Animal , Heterografts , Histone Deacetylases/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Myeloid-Derived Suppressor Cells/drug effects , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects
13.
Neurosci Lett ; 749: 135749, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33610667

ABSTRACT

Exercise is recognized to increase the expression of neurotrophic genes in the hippocampus and prevent cognitive impairment. Histone deacetylase (HDAC) inhibitor acetylate histones and enhance gene transcription in epigenetic regulation. HDAC inhibitors are expected to be an efficacious pharmacological treatment for cognitive function. This study aimed to examine the effect of HDAC inhibitors and exercise on epigenetic markers and neurotrophic gene expression in the hippocampus to find a more enriched brain conditioning for cognitive function based on the synergic effects of pharmacological treatment and behavioral therapy. Thirteen-week-old male mice were divided into four groups. Intraperitoneal administration of an HDAC inhibitor (1.2 g/kg sodium butyrate, NaB) and treadmill exercise (approximately 10 m/min for 60 min) were performed 5 days a week for 4 weeks. NaB administration increased the expression of an immediate-early gene, a neurotrophin, and a neurotrophin receptor in the hippocampus. These results indicate that HDAC inhibition could present an enriched platform for neuronal plasticity in the hippocampus and cognitive function. The novel object recognition test showed that NaB administration increased the score. Notably, the step-through passive avoidance test showed improved learning and memory in the presence of exercise and exercise, indicating that the mice acquired fear memory, specifically in the presence of NaB administration plus exercise. This study found that repetitive administration of HDAC inhibitors improved cognitive function and HDAC inhibitor administration plus exercise has a synergic effect on learning and memory, accompanying the enhancement of crucial gene transcriptions for neuronal plasticity in the hippocampus.


Subject(s)
Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Histone Deacetylases/drug effects , Memory/drug effects , Neuronal Plasticity/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Butyric Acid/pharmacology , Cognition/drug effects , Cognition/physiology , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Male , Mice , Physical Conditioning, Animal/physiology
14.
J Cell Physiol ; 236(8): 5582-5591, 2021 08.
Article in English | MEDLINE | ID: mdl-33481312

ABSTRACT

At present, metabolic diseases, such as obesity and diabetes, have become the world's top health threats. These diseases are closely related to the abnormal development and function of adipocytes and metabolic inflammation associated with obesity. Histone deacetylase 11 (HDAC11), with a relatively unique structure and function in the HDAC family, plays a vital role in regulating cell growth, migration, and cell death. Currently, research on new key regulatory functions of HDAC11 in metabolic homeostasis is receiving more and more attention, and HDAC11 has also become a potential therapeutic target in the treatment of obesity and obesity-related diseases. Here, we summarized the latest literature on the role of HDAC11 in regulating the progress of obesity-related metabolic disorders.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Obesity/drug therapy , Animals , Cell Proliferation/drug effects , Histone Deacetylases/drug effects , Humans , Inflammation/drug therapy
15.
J Pain ; 22(4): 454-471, 2021 04.
Article in English | MEDLINE | ID: mdl-33421591

ABSTRACT

The rationale of spinal administration of endothelin-1(ET-1) mediated anti-nociceptive effect has not been elucidated. ET-1 is reported to promote nuclear effluxion of histone deacetylase 5 (HDAC5) in myocytes, and spinal HDAC5 is implicated in modulation of pain processing. In this study, we aimed to investigate whether central ET-1 plays an anti-nociceptive role by facilitating spinal HDAC5 nuclear shuttling under neuropathic pain. Here, we demonstrate that upregulating spinal ET-1 attenuated the nociception induced by partial sciatic nerve ligation surgery and this analgesic effect mediated by ET-1 was attenuated by intrathecal injection of endothelin A receptor selective inhibitor (BQ123) or by blocking the exportation of nuclear HDAC5 by adeno-associated viruses targeting neuronal HDAC5 (AVV-HDAC5 S259/498A Mutant). Notably, ET-1 administration increased spinal glutamate acid decarboxylases (GAD65/67) expression via initiating HDAC5 nuclear exportation and increased the acetylation of histone 3 at lysine 9 (Acetyl-H3K9) in the promotor regions of spinal Gad1 and Gad2 genes. This was reversed by blocking endothelin A receptor function or by inhibiting the spinal neuronal nuclear exportation of HDAC5. Therefore, inducing spinal GABAergic neuronal HDAC5 nuclear exportation may be a novel therapeutic approach for managing neuropathic pain. PERSPECTIVE: Neuropathic pain is intractable in a clinical setting, and epigenetic regulation is considered to contribute to this processing. Characterizing the anti-nociceptive effect of ET-1 and investigating the associated epigenetic mechanisms in animal models may lead to the development of new therapeutic strategies and targets for treating neuropathic pain.


Subject(s)
Analgesia , Endothelin Receptor Antagonists/pharmacology , Endothelin-1/metabolism , Glutamate Decarboxylase/metabolism , Histone Deacetylases/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Animals , Endothelin Receptor Antagonists/administration & dosage , Endothelin-1/drug effects , Glutamate Decarboxylase/drug effects , Histone Deacetylases/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides, Cyclic/pharmacology
16.
Expert Opin Ther Targets ; 25(1): 75-85, 2021 01.
Article in English | MEDLINE | ID: mdl-33275850

ABSTRACT

Introduction: Epilepsy is a network-level neurological disorder characterized by unprovoked recurrent seizures and associated comorbidities. Aberrant activity and localization of histone deacetylases (HDACs) have been reported in epilepsy and HDAC inhibitors (HDACi) have been used for therapeutic purposes. Several non-histone targets of HDACs have been recognized whose reversible acetylation can modulate protein functions and can contribute to disease pathology. Areas covered: This review provides an overview of HDACs in epilepsy and reflects its action on non-histone substrates involved in the pathogenesis of epilepsy and explores the effectiveness of HDACi as anti-epileptic drugs (AEDs). It also covers the efforts undertaken to target the interaction of HDACs with their substrates. We have further discussed non-deacetylase activity possessed by specific HDACs that might be essential in unraveling the molecular mechanism underlying the disease. For this purpose, relevant literature from 1996 to 2020 was derived from PubMed. Expert opinion: The interaction of HDACs and their non-histone substrates can serve as a promising therapeutic target for epilepsy. Pan-HDACi offers limited benefits to the epileptic patients. Thus, identification of novel targets of HDACs contributing to the disease and designing inhibitors targeting these complexes would be more effective and holds a greater potential as an anti-epileptogenic therapy.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Animals , Anticonvulsants/administration & dosage , Drug Design , Epilepsy/enzymology , Epilepsy/physiopathology , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Molecular Targeted Therapy
17.
Brain Res Bull ; 167: 71-79, 2021 02.
Article in English | MEDLINE | ID: mdl-33232741

ABSTRACT

OBJECTIVE: Hypoxic-ischemic brain damage (HIBD) is a common brain injury caused by hypoxia or ischemia of the brain. This study aims to investigate the effect of dexmedetomidine (Dex) post-treatment on neurological impairment of newborn rats with HIBD via modulating microRNA-29a-3p (miR-29a-3p) and histone deacetylase 4 (HDAC4). METHODS: HIBD model of newborn rats was established. Newborn modeled rats were injected with Dex, miR-29a-3p mimic or HDAC4 siRNA to figure their roles in learning and memory abilities, left hemisphere atrophy, brain tissue injury, inflammatory response and apoptosis rate of nerve cells of rats. The expression of miR-29a-3p and HDAC4 in hippocampal tissues of rats were detected, and the potential relationship between miR-29a-3p and HDAC4 was analyzed. RESULTS: Decreased miR-29a-3p and elevated HDAC4 were found in hippocampal tissues of rats with HIBD. In addition, Dex, elevated miR-29a-3p or declined HDAC4 enhanced spatial learning and memory abilities in rats with HIBD. Moreover, Dex, up-regulated miR-29a-3p or declined HDAC4 alleviated brain atrophy, repressed brain tissue injury, retrained the inflammation, repressed the apoptosis of neurons in the hippocampal region of rats with HIBD. HDAC4 was targeted and negatively regulated by miR-29a-3p. CONCLUSION: The study concludes that miR-29a-3p strengthened the effect of Dex on improving neurologic damage in newborn rats with HIBD by inhibiting HDAC4.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Histone Deacetylases/metabolism , Hypoxia-Ischemia, Brain/pathology , MicroRNAs/metabolism , Animals , Animals, Newborn , Gene Expression Regulation/drug effects , Histone Deacetylases/drug effects , Hypoxia-Ischemia, Brain/metabolism , Male , MicroRNAs/pharmacology , Rats , Rats, Sprague-Dawley
18.
Diabetologia ; 64(1): 226-239, 2021 01.
Article in English | MEDLINE | ID: mdl-33106900

ABSTRACT

AIMS/HYPOTHESIS: Microvascular endothelial hyperpermeability, mainly caused by claudin-5 deficiency, is the initial pathological change that occurs in diabetes-associated cardiovascular disease. The ketone body ß-hydroxybutyrate (BHB) exerts unique beneficial effects on the cardiovascular system, but the involvement of BHB in promoting the generation of claudin-5 to attenuate cardiac microvascular hyperpermeability in diabetes is poorly understood. METHODS: The effects of BHB on cardiac microvascular endothelial hyperpermeability and claudin-5 generation were evaluated in rats with streptozotocin-induced diabetes and in high glucose (HG)-stimulated human cardiac microvascular endothelial cells (HCMECs). To explore the underlying mechanisms, we also measured ß-catenin nuclear translocation, binding of ß-catenin, histone deacetylase (HDAC)1, HDAC3 and p300 to the Claudin-5 (also known as CLDN5) promoter, interaction between HDAC3 and ß-catenin, and histone acetylation in the Claudin-5 promoter. RESULTS: We found that 10 weeks of BHB treatment promoted claudin-5 generation and antagonised cardiac microvascular endothelial hyperpermeability in rat models of diabetes. Meanwhile, BHB promoted claudin-5 generation and inhibited paracellular permeability in HG-stimulated HCMECs. Specifically, BHB (2 mmol/l) inhibited HG-induced HDAC3 from binding to the Claudin-5 promoter, although nuclear translocation or promoter binding of ß-catenin did not change with BHB treatment. In addition, BHB prevented the binding and co-localisation of HDAC3 to ß-catenin in HG-stimulated HCMECs. Furthermore, using mass spectrometry, acetylated H3K14 (H3K14ac) in the Claudin-5 promoter following BHB treatment was identified, regardless of whether cells were stimulated by HG or not. Although reduced levels of acetylated H3K9 in the Claudin-5 promoter were found following HG stimulation, increased H3K14ac was specifically associated with BHB treatment. CONCLUSIONS/INTERPRETATION: BHB inhibited HDAC3 and caused acetylation of H3K14 in the Claudin-5 promoter, thereby promoting claudin-5 generation and antagonising diabetes-associated cardiac microvascular hyperpermeability. Graphical abstract.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Capillary Permeability/drug effects , Claudin-5/biosynthesis , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Histone Deacetylases/drug effects , Animals , Capillary Permeability/physiology , Claudin-5/genetics , Diabetes Complications/prevention & control , Endothelium, Vascular/physiopathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Male , Microvessels/physiopathology , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , beta Catenin/metabolism
19.
Invest Ophthalmol Vis Sci ; 61(14): 4, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33263714

ABSTRACT

Purpose: This study determines if δ-opioid receptor agonist (i.e. SNC-121)-induced epigenetic changes via regulation of histone deacetylases (HDACs) for retinal ganglion cell (RGC) neuroprotection in glaucoma model. Methods: Intraocular pressure was raised in rat eyes by injecting 2M hypertonic saline into the limbal veins. SNC-121 (1 mg/kg; i.p.) was administered to the animals for 7 days. Retinas were collected at days 7 and 42, post-injury followed by measurement of HDAC activities, mRNA, and protein expression by enzyme assay, quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry. Results: The visual acuity, contrast sensitivity, and pattern electroretinograms (ERGs) were declined in ocular hypertensive animals, which were significantly improved by SNC-121 treatment. Class I and IIb HDACs activities were significantly increased at days 7 and 42 in ocular hypertensive animals. The mRNA and protein expression of HDAC 1 was increased by 1.33 ± 0.07-fold and 20.2 ± 2.7%, HDAC 2 by 1.4 ± 0.05-fold and 17.0 ± 2.4%, HDAC 3 by 1.4 ± 0.06-fold and 17.4 ± 3.4%, and HDAC 6 by 1.5 ± 0.09-fold and 15.1 ± 3.3% at day 7, post-injury. Both the mRNA and protein expression of HDACs were potentiated further at day 42 in ocular hypertensive animals. HDAC activities, mRNA, and protein expression were blocked by SNC-121 treatment at days 7 and 42 in ocular hypertensive animals. Conclusions: Data suggests that class I and IIb HDACs are activated and upregulated during early stages of glaucoma. Early intervention with δ-opioid receptor activation resulted in the prolonged suppression of class I and IIb HDACs activities and expression, which may, in part, play a crucial role in RGC neuroprotection.


Subject(s)
Epigenesis, Genetic/drug effects , Glaucoma/metabolism , Histone Deacetylases/metabolism , Receptors, Opioid, delta/agonists , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Female , Glaucoma/enzymology , Histone Deacetylases/drug effects , Intraocular Pressure , Male , Rats , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism
20.
Int J Mol Sci ; 21(22)2020 Nov 22.
Article in English | MEDLINE | ID: mdl-33266366

ABSTRACT

Class I histone deacetylases (HDACs) are promising targets for epigenetic therapies for a range of diseases such as cancers, inflammations, infections and neurological diseases. Although six HDAC inhibitors are now licensed for clinical treatments, they are all pan-inhibitors with little or no HDAC isoform selectivity, exhibiting undesirable side effects. A major issue with the currently available HDAC inhibitors is that they have limited specificity and target multiple deacetylases. Except for HDAC8, Class I HDACs (1, 2 and 3) are recruited to large multiprotein complexes to function. Therefore, there are rising needs to develop new, hopefully, therapeutically efficacious HDAC inhibitors with isoform or complex selectivity. Here, upon the introduction of the structures of Class I HDACs and their complexes, we provide an up-to-date overview of the structure-based discovery of Class I HDAC inhibitors, including pan-, isoform-selective and complex-specific inhibitors, aiming to provide an insight into the discovery of additional HDAC inhibitors with greater selectivity, specificity and therapeutic utility.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Animals , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...