Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Clin Epigenetics ; 16(1): 78, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862980

ABSTRACT

Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the ß-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting ß-cells and reducing T2DM-associated complications, among others.


Subject(s)
Diabetes Mellitus, Type 2 , Epigenesis, Genetic , Histone Deacetylase Inhibitors , Histone Deacetylases , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Oxidative Stress/drug effects , Insulin/metabolism
2.
Adv Sci (Weinh) ; 11(26): e2401939, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704700

ABSTRACT

Obesity is a multifactorial disease that is part of today's epidemic and also increases the risk of other metabolic diseases. Long noncoding RNAs (lncRNAs) provide one tier of regulatory mechanisms to maintain metabolic homeostasis. Although lncRNAs are a significant constituent of the mammalian genome, studies aimed at their metabolic significance, including obesity, are only beginning to be addressed. Here, a developmentally regulated lncRNA, termed as obesity related (Obr), whose expression in metabolically relevant tissues such as skeletal muscle, liver, and pancreas is altered in diet-induced obesity, is identified. The Clone 9 cell line and high-fat diet-induced obese Wistar rats are used as a model system to verify the function of Obr. By using stable expression and antisense oligonucleotide-mediated downregulation of the expression of Obr followed by different molecular biology experiments, its role in lipid metabolism is verified. It is shown that Obr associates with the cAMP response element-binding protein (Creb) and activates different transcription factors involved in lipid metabolism. Its association with the Creb histone acetyltransferase complex, which includes the cAMP response element-binding protein (CBP) and p300, positively regulates the transcription of genes involved in lipid metabolism. In addition, Obr is regulated by Pparγ in response to lipid accumulation.


Subject(s)
Epigenesis, Genetic , Lipid Metabolism , Obesity , RNA, Long Noncoding , Rats, Wistar , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Lipid Metabolism/genetics , Rats , Obesity/genetics , Obesity/metabolism , Epigenesis, Genetic/genetics , Disease Models, Animal , Diet, High-Fat/adverse effects , Male
3.
Cells ; 12(20)2023 10 14.
Article in English | MEDLINE | ID: mdl-37887295

ABSTRACT

BACKGROUND: Genetic and epigenetic changes, oxidative stress and inflammation influence the rate of aging, which diseases, lifestyle and environmental factors can further accelerate. In accelerated aging (AA), the biological age exceeds the chronological age. OBJECTIVE: The objective of this study is to reappraise the AA concept critically, considering its weaknesses and limitations. METHODS: We reviewed more than 300 recent articles dealing with the physiology of brain aging and neurodegeneration pathophysiology. RESULTS: (1) Application of the AA concept to individual organs outside the brain is challenging as organs of different systems age at different rates. (2) There is a need to consider the deceleration of aging due to the potential use of the individual structure-functional reserves. The latter can be restored by pharmacological and/or cognitive therapy, environment, etc. (3) The AA concept lacks both standardised terminology and methodology. (4) Changes in specific molecular biomarkers (MBM) reflect aging-related processes; however, numerous MBM candidates should be validated to consolidate the AA theory. (5) The exact nature of many potential causal factors, biological outcomes and interactions between the former and the latter remain largely unclear. CONCLUSIONS: Although AA is commonly recognised as a perspective theory, it still suffers from a number of gaps and limitations that assume the necessity for an updated AA concept.


Subject(s)
Aging , Oxidative Stress , Humans , Aging/genetics , Epigenesis, Genetic , Brain , Inflammation/genetics , Biomarkers
4.
Cells ; 12(11)2023 05 30.
Article in English | MEDLINE | ID: mdl-37296636

ABSTRACT

Maternal diabetes has been associated with a greater risk of neurodevelopmental disorders in offspring. It has been established that hyperglycemia alters the expression of genes and microRNAs (miRNAs) regulating the fate of neural stem cells (NSCs) during brain development. In this study, the expression of methyl-CpG-binding protein-2 (Mecp2), a global chromatin organizer and a crucial regulator of synaptic proteins, was analyzed in NSCs obtained from the forebrain of embryos of diabetic mice. Mecp2 was significantly downregulated in NSCs derived from embryos of diabetic mice when compared to controls. miRNA target prediction revealed that the miR-26 family could regulate the expression of Mecp2, and further validation confirmed that Mecp2 is a target of miR-26b-5p. Knockdown of Mecp2 or overexpression of miR-26b-5p altered the expression of tau protein and other synaptic proteins, suggesting that miR-26b-5p alters neurite outgrowth and synaptogenesis via Mecp2. This study revealed that maternal diabetes upregulates the expression of miR-26b-5p in NSCs, resulting in downregulation of its target, Mecp2, which in turn perturbs neurite outgrowth and expression of synaptic proteins. Overall, hyperglycemia dysregulates synaptogenesis that may manifest as neurodevelopmental disorders in offspring from diabetic pregnancy.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , MicroRNAs , Neural Stem Cells , Pregnancy , Female , Animals , Mice , Diabetes Mellitus, Experimental/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Hyperglycemia/genetics , Methyl-CpG-Binding Protein 2/genetics
5.
Vitam Horm ; 122: 147-169, 2023.
Article in English | MEDLINE | ID: mdl-36863792

ABSTRACT

Type 2 diabetes (T2D) is a metabolic disease characterized by the development of ß-cell dysfunction with hepatic, muscular and adipose tissue insulin resistance. Although the molecular mechanisms leading to its development are not entirely known, investigations of its causes reveal a multifactorial contribution to its development and progression in most cases. In addition, regulatory interactions mediated by epigenetic modifications such as DNA methylation, histone tail modifications and regulatory RNAs have been found to play a significant role in the etiology of T2D. In this chapter, we discuss the role of DNA methylation and its dynamics in the development of the pathological features of T2D.


Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Adipose Tissue
6.
J Vis Exp ; (183)2022 05 24.
Article in English | MEDLINE | ID: mdl-35695542

ABSTRACT

The current protocol describes the use of lentiviral particles for the delivery of short hairpin RNAs (shRNAs) to both human embryonic stem cells (hESCs) as well as neural progenitor cells (NPCs) derived from hESCs at high efficiency. Lentiviral particles were generated by co-transfecting HEK293T cells using entry vectors (carrying shRNAs) along with packaging plasmids (pAX and pMD2.G) using the low-cost cationic polymer polyethylenimine (PEI). Viral particles were concentrated using ultracentrifugation, which resulted in average titers above 5 x 107. Both hESCs and NPCs could be infected at high efficiencies using these lentiviral particles, as shown by puromycin selection and stable expression in hESCs, as well as transient GFP expression in NPCs. Furthermore, western blot analysis showed a significant reduction in the expression of genes targeted by shRNAs. In addition, the cells retained their pluripotency as well as differentiation potential, as evidenced by their subsequent differentiation into different lineages of CNS. The current protocol deals with the delivery of shRNAs; however, the same approach could be used for the ectopic expression of cDNAs for overexpression studies.


Subject(s)
Human Embryonic Stem Cells , Lentivirus , Genetic Vectors , HEK293 Cells , Humans , Lentivirus/genetics , Polyethyleneimine , Polymers , RNA, Small Interfering/genetics
7.
Open Biol ; 12(4): 210350, 2022 04.
Article in English | MEDLINE | ID: mdl-35382567

ABSTRACT

Although tumourigenesis occurs due to genetic mutations, the role of epigenetic dysregulations in cancer is also well established. Epigenetic dysregulations in cancer may occur as a result of mutations in genes encoding histone/DNA-modifying enzymes and chromatin remodellers or mutations in histone protein itself. It is also true that misregulated gene expression without genetic mutations in these factors could also support tumour initiation and progression. Interestingly, metabolic rewiring has emerged as a hallmark of cancer due to gene mutations in specific metabolic enzymes or dietary/environmental factors. Recent studies report an intricate cross-talk between epigenetic and metabolic reprogramming in cancer. This review discusses the role of epigenetic and metabolic dysregulations and their cross-talk in tumourigenesis with a special focus on gliomagenesis. We also discuss the role of recently developed human embryonic stem cells/induced pluripotent stem cells-derived organoid models of gliomas and how these models are proving instrumental in uncovering human-specific cellular and molecular complexities of gliomagenesis.


Subject(s)
Epigenesis, Genetic , Epigenomics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Chromatin/genetics , Histones/metabolism , Humans
8.
Med Res Rev ; 42(3): 1343-1371, 2022 05.
Article in English | MEDLINE | ID: mdl-34984701

ABSTRACT

A recent International Diabetes Federation report suggests that more than 463 million people between 20 and 79 years have diabetes. Of the 20 million women affected by hyperglycemia during pregnancy, 84% have gestational diabetes. In addition, more than 1.1 million children or adolescents are affected by type 1 diabetes. Factors contributing to the increase in diabetes prevalence are complex and include contributions from genetic, environmental, and epigenetic factors. However, molecular regulatory mechanisms influencing the progression of an individual towards increased susceptibility to metabolic diseases such as diabetes are not fully understood. Recent studies suggest that the pathogenesis of diabetes involves epigenetic changes, resulting in a persistently dysregulated metabolic phenotype. This review summarizes the role of epigenetic mechanisms, mainly DNA methylation and histone modifications, in the development of the pancreas, their contribution to the development of diabetes, and the potential employment of epigenetic modulators in diabetes treatment.


Subject(s)
Diabetes Mellitus, Type 1 , Epigenesis, Genetic , Adolescent , DNA Methylation/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Female , Humans , Pancreas , Phenotype , Pregnancy
9.
J Biol Chem ; 296: 100035, 2021.
Article in English | MEDLINE | ID: mdl-33154167

ABSTRACT

Embryonic and adult stem cells possess the capability of self-renewal and lineage-specific differentiation. The intricate balance between self-renewal and differentiation is governed by developmental signals and cell-type-specific gene regulatory mechanisms. A perturbed intra/extracellular environment during lineage specification could affect stem cell fate decisions resulting in pathology. Growing evidence demonstrates that metabolic pathways govern epigenetic regulation of gene expression during stem cell fate commitment through the utilization of metabolic intermediates or end products of metabolic pathways as substrates for enzymatic histone/DNA modifications. UDP-GlcNAc is one such metabolite that acts as a substrate for enzymatic mono-glycosylation of various nuclear, cytosolic, and mitochondrial proteins on serine/threonine amino acid residues, a process termed protein O-GlcNAcylation. The levels of GlcNAc inside the cells depend on the nutrient availability, especially glucose. Thus, this metabolic sensor could modulate gene expression through O-GlcNAc modification of histones or other proteins in response to metabolic fluctuations. Herein, we review evidence demonstrating how stem cells couple metabolic inputs to gene regulatory pathways through O-GlcNAc-mediated epigenetic/transcriptional regulatory mechanisms to govern self-renewal and lineage-specific differentiation programs. This review will serve as a primer for researchers seeking to better understand how O-GlcNAc influences stemness and may catalyze the discovery of new stem-cell-based therapeutic approaches.


Subject(s)
Cell Lineage , Proteins/metabolism , Stem Cells/cytology , Uridine Diphosphate N-Acetylglucosamine/metabolism , Animals , Epigenesis, Genetic , Gene Regulatory Networks , Humans , Protein Processing, Post-Translational , Stem Cells/metabolism
10.
Clin Epigenetics ; 12(1): 104, 2020 07 11.
Article in English | MEDLINE | ID: mdl-32653024

ABSTRACT

Diabetes mellitus (DM) is a chronic condition characterised by ß cell dysfunction and persistent hyperglycaemia. The disorder can be due to the absence of adequate pancreatic insulin production or a weak cellular response to insulin signalling. Among the three types of DM, namely, type 1 DM (T1DM), type 2 DM (T2DM), and gestational DM (GDM); T2DM accounts for almost 90% of diabetes cases worldwide.Epigenetic traits are stably heritable phenotypes that result from certain changes that affect gene function without altering the gene sequence. While epigenetic traits are considered reversible modifications, they can be inherited mitotically and meiotically. In addition, epigenetic traits can randomly arise in response to environmental factors or certain genetic mutations or lesions, such as those affecting the enzymes that catalyse the epigenetic modification. In this review, we focus on the role of DNA methylation, a type of epigenetic modification, in the pathogenesis of T2DM.


Subject(s)
DNA Methylation/genetics , Diabetes Mellitus, Type 2/genetics , Insulin/genetics , Mutation/genetics , Adult , Aged , Animals , Case-Control Studies , CpG Islands/genetics , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Environment , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Gene Expression , Humans , Insulin/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Prospective Studies , Rats , Repressor Proteins/genetics , Transcriptional Activation/genetics
11.
Dis Model Mech ; 11(3)2018 03 01.
Article in English | MEDLINE | ID: mdl-29434026

ABSTRACT

Although the existence of a close relationship between the early maternal developmental environment, fetal size at birth and the risk of developing disease in adulthood has been suggested, most studies, however, employed experimentally induced intrauterine growth restriction as a model to link this with later adult disease. Because embryonic size variation also occurs under normal growth and differentiation, elucidating the molecular mechanisms underlying these changes and their relevance to later adult disease risk becomes important. The birth weight of rat pups vary according to the uterine horn positions. Using birth weight as a marker, we compared two groups of rat pups - lower birth weight (LBW, 5th to 25th percentile) and average birth weight (ABW, 50th to 75th percentile) - using morphological, biochemical and molecular biology, and genetic techniques. Our results show that insulin metabolism, Pi3k/Akt and Pparγ signaling and the genes regulating growth and metabolism are significantly different in these groups. Methylation at the promoter of the InsII (Ins2) gene and DNA methyltransferase 1 in LBW pups are both increased. Additionally, the Dnmt1 repressor complex, which includes Hdac1, Rb (Rb1) and E2f1, was also upregulated in LBW pups. We conclude that the Dnmt1 repressor complex, which regulates the restriction point of the cell cycle, retards the rate at which cells traverse the G1 or G0 phase of the cell cycle in LBW pups, thereby slowing down growth. This regulatory mechanism mediated by Dnmt1 might contribute to the production of small-size pups and altered physiology and pathology in adult life.


Subject(s)
Growth and Development , Metabolism , Animals , Animals, Newborn , Birth Weight , Cell Cycle/genetics , DNA Methylation/genetics , Fatty Acids/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glucagon/metabolism , Glucose/metabolism , Growth and Development/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Metabolism/genetics , Methyltransferases/metabolism , Models, Animal , Muscle, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction
12.
Biochim Biophys Acta ; 1779(10): 634-44, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18723134

ABSTRACT

The expression of PR-1a gene in tobacco is accompanied by changes in the chromatin architecture over its promoter region. The transcription initiates when the gene is induced in defense response, a condition that can be simulated experimentally by external application of salicylic acid. Mutagenesis of the core promoter sequence established that the TATA-box was critical to the expression of PR-1a gene. In order to study functional specificity between the core promoter and upstream activator region, the native core promoter was exchanged with that of a heterologous salicylic acid inducible promoter, Pcec. The core promoter and the activator region of PR-1a together determine its tightly regulated expression, slow kinetics of induction by SA and several fold induction of expression. In uninduced state, a single nucleosome was present over the core promoter of PR-1a. It masked both the TATA-box and the transcription initiation region. The transcriptional activation of the promoter by SA was accompanied by shift in the position of this nucleosome. The chimeric promoters failed to show inducibility or gave very low level of induction. They showed failure in shifting the nucleosome from the core promoter region. The promoter Pcec expressed constitutively at a high uninduced level in spite of a nucleosome over the TATA-box region. However, in this case, the nucleosome did not mask the transcript initiation region. The TATA-box nucleosome was shifted as the expression increased further, following induction by SA. A fully induced Pcec had the TATA-box fully exposed, though a weak nucleosome appeared on the +1 region. The results support a close relationship among promoter sequence architecture, nucleosome positioning and PR-1a expression.


Subject(s)
Epistasis, Genetic/physiology , Gene Expression Regulation, Plant , Nicotiana/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Base Sequence/physiology , Gene Expression Regulation, Plant/drug effects , Genes, Plant , Host-Pathogen Interactions/genetics , Models, Biological , N-Acetylneuraminic Acid/pharmacology , TATA Box/physiology
13.
J Biotechnol ; 123(1): 1-12, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16324763

ABSTRACT

A synthetic bidirectional expression module was constructed by placing a computationally designed minimal promoter sequence on the 5' and 3' sides of a transcription activation module. The activation of transcription from the unidirectional and bidirectional promoters constructed from the same sequence elements was evaluated by using the reporter genes gusA and gfp. The analysis based on transient and stable transformation of tobacco showed that the artificially designed multifactorial activation module activated transcription simultaneously to comparable levels in both the directions. The transcription activation module responded to elicitors like salicylic acid, NaCl and IAA in the forward as well as reverse directions. The concentration of the elicitor required for highest gene activation was similar for the two directions in case of the three activators. The kinetics of time of induction was similar in the two directions for salicylic acid and NaCl. In the case of IAA, the transcription activation was faster in the reverse direction. The results show that constitutive and chemically inducible bidirectional promoters can be deployed for predictable simultaneous regulation of two genes for genetic engineering in plants.


Subject(s)
Gene Expression/physiology , Nicotiana/genetics , Plant Proteins/genetics , Plants, Genetically Modified/metabolism , Promoter Regions, Genetic/genetics , Protein Engineering/methods , Transgenes/genetics , Factor Analysis, Statistical , Gene Expression Regulation, Plant/genetics , Genetic Enhancement/methods , Plant Proteins/metabolism , Sequence Analysis, DNA/methods , Nicotiana/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...