Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Aging Cell ; 23(5): e14108, 2024 May.
Article in English | MEDLINE | ID: mdl-38408164

ABSTRACT

Histones serve as a major carrier of epigenetic information in the form of post-translational modifications which are vital for controlling gene expression, maintaining cell identity, and ensuring proper cellular function. Loss of histones in the aging genome can drastically impact the epigenetic landscape of the cell leading to altered chromatin structure and changes in gene expression profiles. In this study, we investigated the impact of age-related changes on histone levels and histone acetylation in the retinal pigment epithelium (RPE) and retina of mice. We observed a global reduction of histones H1, H2A, H2B, H3, and H4 in aged RPE/choroid but not in the neural retina. Transcriptomic analyses revealed significant downregulation of histones in aged RPE/choroid including crucial elements of the histone locus body (HLB) complex involved in histone pre-mRNA processing. Knockdown of HINFP, a key HLB component, in human RPE cells induced histone loss, senescence, and the upregulation of senescence-associated secretory phenotype (SASP) markers. Replicative senescence and chronological aging in human RPE cells similarly resulted in progressive histone loss and acquisition of the SASP. Immunostaining of human retina sections revealed histone loss in RPE with age. Acetyl-histone profiling in aged mouse RPE/choroid revealed a specific molecular signature with loss of global acetyl-histone levels, including H3K14ac, H3K56ac, and H4K16ac marks. These findings strongly demonstrate histone loss as a unique feature of RPE aging and provide critical insights into the potential mechanisms linking histone dynamics, cellular senescence, and aging.


Subject(s)
Aging , Histones , Retinal Pigment Epithelium , Retinal Pigment Epithelium/metabolism , Histones/metabolism , Animals , Acetylation , Mice , Aging/metabolism , Humans , Cellular Senescence , Mice, Inbred C57BL
2.
Article in English | MEDLINE | ID: mdl-37937078

ABSTRACT

Introduction: Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis, cytopenias, and dysplasia. The gene encoding ten-eleven translocation 2 (tet2), a dioxygenase enzyme that catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine, is a recurrently mutated tumor suppressor gene in MDS and other myeloid malignancies. Previously, we reported a stable zebrafish line with a loss-of-function mutation in the tet2 gene. The tet2m/m-mutant zebrafish developed a pre-MDS state with kidney marrow dysplasia, but normal circulating blood counts by 11 months of age and accompanying anemia, signifying the onset of MDS, by 24 months of age. Methods: In the current study, we collected progenitor cells from the kidney marrows of the adult tet2m/m and tet2wt/wt fish at 4 and 15 months of age and conducted enhanced reduced representation of bisulfite sequencing (ERRBS) and bulk RNA-seq to measure changes in DNA methylation and gene expression of hematopoietic stem and progenitor cells (HSPCs). Results and discussion: A global increase in DNA methylation of gene promoter regions and CpG islands was observed in tet2m/m HSPCs at 4 months of age when compared with the wild type. Furthermore, hypermethylated genes were significantly enriched for targets of SUZ12 and the metal-response-element-binding transcription factor 2 (MTF2)-involved in the polycomb repressive complex 2 (PRC2). However, between 4 and 15 months of age, we observed a paradoxical global decrease in DNA methylation in tet2m/m HSPCs. Gene expression analyses identified upregulation of genes associated with mTORC1 signaling and interferon gamma and alpha responses in tet2m/m HSPCs at 4 months of age when compared with the wild type. Downregulated genes in HSPCs of tet2-mutant fish at 4 months of age were enriched for cell cycle regulation, heme metabolism, and interleukin 2 (IL2)/signal transducer and activator of transcription 5 (STAT5) signaling, possibly related to increased self-renewal and clonal advantage in HSPCs with tet2 loss of function. Finally, there was an overall inverse correlation between overall increased promoter methylation and gene expression.

3.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982266

ABSTRACT

The CRISPR/Cas9 system is a robust, efficient, and cost-effective gene editing tool widely adopted in translational studies of ocular diseases. However, in vivo CRISPR-based editing in animal models poses challenges such as the efficient delivery of the CRISPR components in viral vectors with limited packaging capacity and a Cas9-associated immune response. Using a germline Cas9-expressing mouse model would help to overcome these limitations. Here, we evaluated the long-term effects of SpCas9 expression on retinal morphology and function using Rosa26-Cas9 knock-in mice. We observed abundant SpCas9 expression in the RPE and retina of Rosa26-Cas9 mice using the real-time polymerase chain reaction (RT-PCR), Western blotting, and immunostaining. SD-OCT imaging and histological analysis of the RPE, retinal layers, and vasculature showed no apparent structural abnormalities in adult and aged Cas9 mice. Full-field electroretinogram of adult and aged Cas9 mice showed no long-term functional changes in the retinal tissues because of constitutive Cas9 expression. The current study showed that both the retina and RPE maintain their phenotypic and functional features in Cas9 knock-in mice, establishing this as an ideal animal model for developing therapeutics for retinal diseases.


Subject(s)
CRISPR-Cas Systems , Retina , Mice , Animals , Retina/metabolism , Gene Editing/methods , Electroretinography , Genetic Vectors
4.
Biotechniques ; 72(3): 81-84, 2022 03.
Article in English | MEDLINE | ID: mdl-35119307

ABSTRACT

Acute myeloid leukemia patients with FMS-like tyrosine kinase 3-internal tandem duplications and mixed lineage leukemia-protein AF9 fusion proteins suffer from poor clinical outcomes. The MOLM-13 acute myeloid leukemia cell line harbors both of these abnormalities and is used in CRISPR experiments to identify disease drivers. However, experimental observations may be biased or inconclusive in the absence of experimentally validated positive control genes. We validated sgRNAs for knockdown of TP53 for cell proliferation and for DCK knockdown and CDA upregulation for cytarabine resistance control genes in MOLM-13 cells. We have provided a detailed CRISPR protocol applicable to both gene knockdown or activation experiments and downstream leukemic phenotype analyses. Inclusion of these controls in CRISPR experiments will enhance the capacity to identify novel myeloid leukemia drivers in MOLM-13 cells.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cytarabine/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics
5.
Mol Biol Rep ; 43(6): 457-62, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27153830

ABSTRACT

Dipeptidyl peptidase III (DPP III) is an emerging biomarker of human cancers. Expression, specificity, and function of human DPP III (hDPP III) mRNA variant I (V-I), II (V-II), and III (V-III) are poorly understood. Here, we investigated expression of these variants in multiple human tumor derived cell lines. DNA sequencing revealed concurrent expression of hDPP III V-I and V-II in U87MG (glioblastoma), SCC4 (squamous cell carcinoma), SiHa (carcinoma of uterus) cells. In SKOV1 cells, a cell line derived from ovarian carcinoma where a positive correlation between histological aggressiveness of the malignancy and hDPP III expression has previously been established, only V-II could be detected. Human DPP III V-III, which lacks an in-frame coding sequence, could not be detected in any of these cell lines. 5' untranslated region (UTR) of hDPP III V-II contains nucleotides GCA (-12 to -10 bp) upstream to the translation initiator codon (AUG). These nucleotides are absent from V-I and V-III, however, both V-I and V-II encode for the same hDPP III protein isoform-I. In vitro transcription coupled translation assay using hDPP III V-I and V-II expression vectors which contained full length V-I and V-II cDNA including the variable 5' UTR cloned under T7 promoter, respectively revealed a comparable translational efficiency for both the variants, abrogating involvement of nucleotides GCA (-12 to -10 bp) in translation of the variants. Our results, for the first time, demonstrate concurrent expression in multiple tumor derived cell lines and a comparable in vitro translational efficiency for hDPP III V-I and II.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Cell Line, Tumor , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Biosynthesis
6.
Biol Chem ; 397(6): 563-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26887037

ABSTRACT

The precise biological function of human dipeptidyl peptidase III (hDPP III) is poorly understood. Using luciferase reporter constructs responsive to change in Ca2+ and/or cAMP and Fura 2-AM fluorometric assay, we show a significant decrease in intracellular Ca2+ following hDPP III overexpression and angiotensin II stimulation in angiotensin II type 1 receptor (G-protein coupled receptor, GPCR) expressing HEK293T cells. Silencing the expression of hDPP III by siRNA reversed the effect of hDPP III overexpression with a concomitant increase in Ca2+. These results, for the first time, show involvement of hDPP III in GPCR dependent Ca2+ regulation in HEK293T cells.


Subject(s)
Calcium/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Receptors, G-Protein-Coupled/metabolism , Cyclic AMP/metabolism , HEK293 Cells , Humans , Intracellular Space/metabolism
7.
FEBS J ; 278(18): 3256-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21794094

ABSTRACT

Dipeptidyl peptidase III (DPP III), the sole member and representative of the M49 family of metallopeptidases, is a zinc-dependent aminopeptidase. It sequentially hydrolyses dipeptides from the N-terminal of oligopeptides ranging from three to 10 amino acid residues. Although implicated in an array of pathophysiological phenomena, the precise function of this peptidase is still unclear. However, a number of studies advocate its contribution in terminal stages of protein turnover. Altered expression of DPP III which suggests its involvement in primary ovarian carcinoma, oxidative stress (Nrf2 nuclear localization), pain, inflammation and cataractogenesis has recently led to resurgence of interest in delineating the role of the peptidase in these pathophysiological processes. This review article intends to bring forth the latest updates in this arena which may serve as a base for future studies on the peptidase.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Oligopeptides/metabolism , Animals , Biocatalysis , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Enzyme Activation , Humans , Metalloexopeptidases/chemistry , Metalloexopeptidases/metabolism , Oxidative Stress , Protease Inhibitors , Protein Conformation , Protein Transport , Substrate Specificity , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...