Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
ACS Appl Bio Mater ; 7(3): 1478-1489, 2024 03 18.
Article in English | MEDLINE | ID: mdl-38354406

ABSTRACT

A major obstacle to axonal regeneration following spinal cord injury (SCI) is neuroinflammation mediated by astrocytes and microglial cells. We previously demonstrated that graphene-based collagen hydrogels alone can decrease neuroinflammation in SCI. Their regenerative potential, however, is poorly understood and incomplete. Furthermore, stem cells have demonstrated both neuroprotective and regenerative properties in spinal cord regeneration, although there are constraints connected with the application of stem cell-based therapy. In this study, we have analyzed the regeneration capability of human bone marrow mesenchymal stem cell (BM-MSC)-loaded graphene-cross-linked collagen cryogels (Gr-Col) in a thoracic (T10-T11) hemisection model of SCI. Our study found that BM-MSC-loaded Gr-Col improves axonal regeneration, reduces neuroinflammation by decreasing astrocyte reactivity, and promotes M2 macrophage polarization. BM-MSC-loaded-Gr-Col demonstrated enhanced regenerative potential compared to Gr-Col and the injury group control. Next-generation sequencing (NGS) analysis revealed that BM-MSC-loaded-Gr-Col modulates the JAK2-STAT3 pathway, thus decreasing the reactive and scar-forming astrocyte phenotype. The decrease in neuroinflammation in the BM-MSC-loaded-Gr-Col group is attributed to the modulation of Notch/Rock and STAT5a/b and STAT6 signaling. Overall, Gene Set Enrichment Analysis suggests the promising role of BM-MSC-loaded-Gr-Col in promoting axonal regeneration after SCI by modulating molecular pathways such as the PI3/Akt pathway, focal adhesion kinase, and various inflammatory pathways.


Subject(s)
Graphite , Mesenchymal Stem Cells , Spinal Cord Injuries , Rats , Animals , Humans , Cryogels/metabolism , Neuroinflammatory Diseases , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Collagen , Mesenchymal Stem Cells/metabolism
2.
Bioorg Med Chem ; 81: 117193, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36796126

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common malignant epithelial neoplasm, affects the mouth and throat, and accounts for 90 % of oral cancers. Considering the associated morbidity with neck dissections and the limitation of existing therapeutic agents, the discovery and development of new anticancer drugs/drug candidates for oral cancer treatment are of the utmost need. In this context, reported here is the identification of fluorinated 2­styryl 4(3H)-quinazolinone as a promising hit for oral cancer. Preliminary studies indicate that the compound blocks the transition of G1 to S phase, thereby leading to arrest in the G1/S phase. Subsequent RNA-seq analysis revealed that the compound induces the activation of molecular pathways involved in apoptosis (such as TNF signalling through NF-κB, p53 pathways) and cell differentiation and suppresses the pathways of cellular growth and development (such as KRAS signaling) in CAL-27 cancer cells. It is noted that identified hit complies with a favorable range of ADME properties as per the computational analysis.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Humans , Apoptosis , Cell Line, Tumor , Cell Proliferation , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Signal Transduction
3.
Life Sci Alliance ; 2(1): e201800115, 2019 02.
Article in English | MEDLINE | ID: mdl-30620009

ABSTRACT

Chromatin immunoprecipitation (ChIP)-seq analyses of transcription factors in clinical specimens are challenging due to the technical limitations and low quantities of starting material, often resulting in low enrichments and poor signal-to-noise ratio. Here, we present an optimized protocol for transcription factor ChIP-seq analyses in human tissue, yielding an ∼100% success rate for all transcription factors analyzed. As proof of concept and to illustrate general applicability of the approach, human tissue from the breast, prostate, and endometrial cancers were analyzed. In addition to standard formaldehyde fixation, disuccinimidyl glutarate was included in the procedure, greatly increasing data quality. To illustrate the sensitivity of the optimized protocol, we provide high-quality ChIP-seq data for three independent factors (AR, FOXA1, and H3K27ac) from a single core needle prostate cancer biopsy specimen. In summary, double-cross-linking strongly improved transcription factor ChIP-seq quality on human tumor samples, further facilitating and enhancing translational research on limited amounts of tissue.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Base Sequence/genetics , Binding Sites/genetics , Biopsy, Large-Core Needle , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Data Accuracy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Hepatocyte Nuclear Factor 3-alpha/genetics , Histones/genetics , Humans , MCF-7 Cells , Male , Receptors, Androgen/genetics , Sensitivity and Specificity
4.
Cell Rep ; 26(4): 1059-1069.e6, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673601

ABSTRACT

Global investigation of histone marks in acute myeloid leukemia (AML) remains limited. Analyses of 38 AML samples through integrated transcriptional and chromatin mark analysis exposes 2 major subtypes. One subtype is dominated by patients with NPM1 mutations or MLL-fusion genes, shows activation of the regulatory pathways involving HOX-family genes as targets, and displays high self-renewal capacity and stemness. The second subtype is enriched for RUNX1 or spliceosome mutations, suggesting potential interplay between the 2 aberrations, and mainly depends on IRF family regulators. Cellular consequences in prognosis predict a relatively worse outcome for the first subtype. Our integrated profiling establishes a rich resource to probe AML subtypes on the basis of expression and chromatin data.


Subject(s)
Chromatin , Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , Mutation , Nuclear Proteins , Oncogene Proteins, Fusion , Chromatin/genetics , Chromatin/metabolism , Chromatin/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
5.
Sci Rep ; 8(1): 15454, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337563

ABSTRACT

Despite the fact that loss of E-cadherin is causal to the development and progression of invasive lobular carcinoma (ILC), options to treat this major breast cancer subtype are limited if tumours develop resistance to anti-oestrogen treatment regimens. This study aimed to identify clinically targetable pathways that are aberrantly active downstream of E-cadherin loss in ILC. Using a combination of reverse-phase protein array (RPPA) analyses, mRNA sequencing, conditioned medium growth assays and CRISPR/Cas9-based knock-out experiments, we demonstrate that E-cadherin loss causes increased responsiveness to autocrine growth factor receptor (GFR)-dependent activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt signalling. Autocrine activation of GFR signalling and its downstream PI3K/Akt hub was independent of oncogenic mutations in PIK3CA, AKT1 or PTEN. Analyses of human ILC samples confirmed growth factor production and pathway activity. Pharmacological inhibition of Akt using AZD5363 or MK2206 resulted in robust inhibition of cell growth and survival of ILC cells, and impeded tumour growth in a mouse ILC model. Because E-cadherin loss evokes hypersensitisation of PI3K/Akt activation independent of oncogenic mutations in this pathway, we propose clinical intervention of PI3K/Akt in ILC based on functional E-cadherin inactivation, irrespective of activating pathway mutations.


Subject(s)
Autocrine Communication , Breast Neoplasms/metabolism , Cadherins/deficiency , Carcinoma, Lobular/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , MCF-7 Cells , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology
6.
Oncotarget ; 9(39): 25630-25646, 2018 May 22.
Article in English | MEDLINE | ID: mdl-29876013

ABSTRACT

Epigenetic alterations have been associated with both pathogenesis and progression of cancer. By screening of library compounds, we identified a novel hybrid epi-drug MC2884, a HAT/EZH2 inhibitor, able to induce bona fide cancer-selective cell death in both solid and hematological cancers in vitro, ex vivo and in vivo xenograft models. Anticancer action was due to an epigenome modulation by H3K27me3, H3K27ac, H3K9/14ac decrease, and to caspase-dependent apoptosis induction. MC2884 triggered mitochondrial pathway apoptosis by up-regulation of cleaved-BID, and strong down-regulation of BCL2. Even aggressive models of cancer, such as p53-/- or TET2-/- cells, responded to MC2884, suggesting MC2884 therapeutic potential also for the therapy of TP53 or TET2-deficient human cancers. MC2884 induced massive apoptosis in ex vivo human primary leukemia blasts with poor prognosis in vivo, by targeting BCL2 expression. MC2884-treatment reduced acetylation of the BCL2 promoter at higher level than combined p300 and EZH2 inhibition. This suggests a key role for BCL-2 reduction in potentiating responsiveness, also in combination therapy with BCL2 inhibitors. Finally, we identified both the mechanism of MC2884 action as well as a potential therapeutic scheme of its use. Altogether, this provides proof of concept for the use of epi-drugs coupled with epigenome analyses to 'personalize' precision medicine.

7.
Oncotarget ; 9(39): 25647-25660, 2018 May 22.
Article in English | MEDLINE | ID: mdl-29876014

ABSTRACT

Epigenomic alterations have been associated with both pathogenesis and progression of cancer. Here, we analyzed the epigenome of two high-risk APL (hrAPL) patients and compared it to non-high-risk APL cases. Despite the lack of common genetic signatures, we found that human hrAPL blasts from patients with extremely poor prognosis display specific patterns of histone H3 acetylation, specifically hyperacetylation at a common set of enhancer regions. In addition, unique profiles of the repressive marks H3K27me3 and DNA methylation were exposed in high-risk APLs. Epigenetic comparison with low/intermediate-risk APLs and AMLs revealed hrAPL-specific patterns of histone acetylation and DNA methylation, suggesting these could be further developed into markers for clinical identification. The epigenetic drug MC2884, a newly generated general HAT/EZH2 inhibitor, induces apoptosis of high-risk APL blasts and reshapes their epigenomes by targeting both active and repressive marks. Together, our analysis uncovers distinctive epigenome signatures of hrAPL patients, and provides proof of concept for use of epigenome profiling coupled to epigenetic drugs to 'personalize' precision medicine.

8.
Sci Rep ; 7(1): 10801, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883544

ABSTRACT

Fibroblast (Fb) differentiation and interstitial fibrosis contribute to cardiac remodeling and loss of function after myocardial infarction (MI). We investigated regional presence and regulation of fibrosis in a pig MI model. In vivo analysis of regional function and perfusion defined three regions: the scar, the myocardium adjacent to the scar (MIadjacent, reduced function, reduced perfusion reserve), and the remote myocardium (MIremote, minimal functional deficit, maintained perfusion). Interstitial and perivascular fibrosis, and increase of collagen type I, was only observed in the MIadjacent. Fb activated protein-alpha (FAP-α) was enriched in MIadjacent compared to MIremote. TGF-ß1, which triggers Fb differentiation, was upregulated in both MIadjacent and MIremote, whereas lysyl oxidase, a regulator of collagen cross-linking, and the proteoglycans decorin and biglycan were only increased in the MIadjacent. Fb isolated and cultured for 4 days had myoFb characteristics with little difference between MIremote and MIadjacent, although RNA sequencing revealed differences in gene expression profiles. Fbs from all regions maintained proliferative capacity, and induced contraction of 3-D collagen matrices but scar myoFb was more effective. These data suggest that after MI, signaling through TGF-ß1, possibly related to increased mechanical load, drives Fb activation throughout the left ventricle while regional signaling determines further maturation and extracellular matrix remodeling after MI.


Subject(s)
Cell Differentiation , Fibroblasts/physiology , Fibrosis/pathology , Heart Ventricles/pathology , Myocardial Infarction/complications , Myocardial Infarction/pathology , Animals , Collagen Type I/metabolism , Disease Models, Animal , Endopeptidases , Gelatinases/metabolism , Membrane Proteins/metabolism , Protein-Lysine 6-Oxidase/metabolism , Proteoglycans/metabolism , Serine Endopeptidases/metabolism , Signal Transduction , Stress, Mechanical , Swine , Transforming Growth Factor beta1/metabolism
9.
Oncotarget ; 8(8): 12855-12865, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28030795

ABSTRACT

Chromosomal translocations are one of the hallmarks of acute myeloid leukemia (AML), often leading to gene fusions and expression of an oncofusion protein. Over recent years it has become clear that most of the AML associated oncofusion proteins molecularly adopt distinct mechanisms for inducing leukemogenesis. Still these unique molecular properties of the chimeric proteins converge and give rise to a common pathogenic molecular mechanism. In the present study we compared genome-wide DNA binding and transcriptome data associated with AML1-ETO, CBFB-MYH11 and PML-RARA oncofusion protein expression to identify unique and common features. Our analyses revealed targeting of oncofusion binding sites to RUNX1 and ETS-factor occupied genomic regions. In addition, it revealed a highly comparable global histone acetylation pattern, similar expression of common target genes and related enrichment of several biological pathways critical for maintenance of AML, suggesting oncofusion proteins deregulate common gene programs despite their distinct binding signatures and mechanisms of action.


Subject(s)
Gene Expression Regulation, Leukemic/physiology , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Acetylation , Carcinogenesis/genetics , Carcinogenesis/metabolism , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-ets-1/genetics , RUNX1 Translocation Partner 1 Protein , Transcriptome
10.
Cell Rep ; 17(8): 2087-2100, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851970

ABSTRACT

The t(8;21) acute myeloid leukemia (AML)-associated oncoprotein AML1-ETO disrupts normal hematopoietic differentiation. Here, we have investigated its effects on the transcriptome and epigenome in t(8,21) patient cells. AML1-ETO binding was found at promoter regions of active genes with high levels of histone acetylation but also at distal elements characterized by low acetylation levels and binding of the hematopoietic transcription factors LYL1 and LMO2. In contrast, ERG, FLI1, TAL1, and RUNX1 bind at all AML1-ETO-occupied regulatory regions, including those of the AML1-ETO gene itself, suggesting their involvement in regulating AML1-ETO expression levels. While expression of AML1-ETO in myeloid differentiated induced pluripotent stem cells (iPSCs) induces leukemic characteristics, overexpression increases cell death. We find that expression of wild-type transcription factors RUNX1 and ERG in AML is required to prevent this oncogene overexpression. Together our results show that the interplay of the epigenome and transcription factors prevents apoptosis in t(8;21) AML cells.


Subject(s)
Apoptosis/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Translocation, Genetic , Acetylation , Base Sequence , Cell Line, Tumor , Cell Lineage/genetics , Cell Survival/genetics , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Gene Knockdown Techniques , Genome, Human , Histone Deacetylases/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Oncogenes , Promoter Regions, Genetic , Protein Binding/genetics , Transcriptional Regulator ERG/metabolism
11.
Exp Hematol ; 42(8): 637-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24814246

ABSTRACT

Transcription factors control expression of genes essential for the normal functioning of the hematopoietic system and regulate development of distinct blood cell types. During leukemogenesis, aberrant regulation of transcription factors such as RUNX1, CBFß, MLL, C/EBPα, SPI1, GATA, and TAL1 is central to the disease. Here, we will discuss the mechanisms of transcription factor deregulation in leukemia and how in recent years next-generation sequencing approaches have helped to elucidate the molecular role of many of these aberrantly expressed transcription factors. We will focus on the complexes in which these factors reside, the role of posttranslational modification of these factors, their involvement in setting up higher order chromatin structures, and their influence on the local epigenetic environment. We suggest that only comprehensive knowledge on all these aspects will increase our understanding of aberrant gene expression in leukemia as well as open new entry points for therapeutic intervention.


Subject(s)
Leukemia/genetics , Transcription Factors/genetics , Gene Expression Regulation, Leukemic , Hematopoiesis , Humans
12.
Blood ; 120(19): 4038-48, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22983443

ABSTRACT

ERG and FLI1 are closely related members of the ETS family of transcription factors and have been identified as essential factors for the function and maintenance of normal hematopoietic stem cells. Here genome-wide analysis revealed that both ERG and FLI1 occupy similar genomic regions as AML1-ETO in t(8;21) AMLs and identified ERG/FLI1 as proteins that facilitate binding of oncofusion protein complexes. In addition, we demonstrate that ERG and FLI1 bind the RUNX1 promoter and that shRNA-mediated silencing of ERG leads to reduced expression of RUNX1 and AML1-ETO, consistent with a role of ERG in transcriptional activation of these proteins. Finally, we identify H3 acetylation as the epigenetic mark preferentially associated with ETS factor binding. This intimate connection between ERG/FLI1 binding and H3 acetylation implies that one of the molecular strategies of oncofusion proteins, such as AML1-ETO and PML-RAR-α, involves the targeting of histone deacetylase activities to ERG/FLI1 bound hematopoietic regulatory sites. Together, these results highlight the dual importance of ETS factors in t(8;21) leukemogenesis, both as transcriptional regulators of the oncofusion protein itself as well as proteins that facilitate AML1-ETO binding.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Trans-Activators/metabolism , Acetylation , Antigens, CD34/metabolism , Binding Sites , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Nucleotide Motifs , Protein Binding , Proto-Oncogene Proteins c-ets/metabolism , RNA Interference , RUNX1 Translocation Partner 1 Protein , Transcriptional Regulator ERG , Translocation, Genetic
13.
3 Biotech ; 1(1): 47-56, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22558535

ABSTRACT

A computational approach for identifying functionally relevant SNPs in gene LIG1 has been proposed. LIG1 is a crucial gene which is involved in excision repair pathways and mutations in this gene may lead to increase sensitivity towards DNA damaging agents. A total of 792 SNPs were reported to be associated with gene LIG1 in dbSNP. Different web server namely SIFT, PolyPhen, CUPSAT, FASTSNP, MAPPER and dbSMR were used to identify potentially functional SNPs in gene LIG1. SIFT, PolyPhen and CUPSAT servers predicted eleven nsSNPs to be intolerant, thirteen nsSNP to be damaging and two nsSNPs have the potential to destabilize protein structure. The nsSNP rs11666150 was predicted to be damaging by all three servers and its mutant structure showed significant increase in overall energy. FASTSNP predicted twenty SNPs to be present in splicing modifier binding sites while rSNP module from MAPPER server predicted nine SNPs to influence the binding of transcription factors. The results from the study may provide vital clues in establishing affect of polymorphism on phenotype and in elucidating drug response.

SELECTION OF CITATIONS
SEARCH DETAIL
...