Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Prog Orthod ; 22(1): 33, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34657984

ABSTRACT

OBJECTIVES: Literature shows that the expression of various biomarkers in peri-miniscrew crevicular fluid (PMICF) is related to the stability of miniscrew implants (MSIs). The present study investigated the role and alterations in levels of circulating cell-free nucleic acids (cfNAs) in PMICF before and after orthodontic loading. MATERIAL AND METHODS: This prospective study consisted of forty-six MSIs placed between the second premolar and first molar in the maxillary and mandibular arches. Direct loading was done after 3 weeks of MSI insertion with nickel-titanium closed coil spring exerting a force of 200 g. The PMICF sample was collected at various time intervals, and the level of cfNA was determined. Clinical parameters, including implant mobility and gingival health, were also assessed. Pre-loading and post-loading parameters were assessed using Wilcoxon's rank-sum test. RESULTS: Among 46 MSIs, 36 were stable during the study and 10 MSIs showed peri-implant inflammation and increased mobility. There was a significant rise in the cfNA concentration 24 h after implant insertion (0.4 ± 0.86 ng/µl). The level of cfNAs significantly decreased over 3 weeks and reached the baseline level (0.2 ± 0.31 ng/µl). There was also a significant rise in the levels of cfNA (0.8 ± 0.70 ng/µl) at 24 h after loading MSIs, which gradually decreased to 0.2 ± 0.24 ng/µl after 63 days. The expression of cfNAs was on the average 0.32 units more in the cases with failed implants (P = 0.05). CONCLUSIONS: cfNA levels in PMICF showed an upward trend 24 h after MSI insertion and 24 h after orthodontic loading. The expression of cfNA was more in cases with failed MSIs. Hence, the cfNAs can be considered as a prognostic biomarker of MSI stability.


Subject(s)
Cell-Free Nucleic Acids , Orthodontic Anchorage Procedures , Bone Screws , Humans , Prospective Studies , Tooth Movement Techniques
2.
Int Orthod ; 19(3): 415-424, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34281788

ABSTRACT

BACKGROUND: Micro-osteoperforation (MOP), a minimally invasive technique for accelerating the rate of orthodontic tooth movement has been research extensively, but with varied clinical results. OBJECTIVE: To compare the efficacy of one-time versus two-time micro-osteoperforation on the rate of maxillary canine retraction, its influence on anchorage loss, canine angulation and the levels of interleukin (IL-1ß) in gingival crevicular fluid (GCF). MATERIALS AND METHODS: The split-mouth study included 16 patients in which the left and right sides were randomly allocated to the control side (one-time MOP) and experimental side (two-time MOP). MOP was performed on both sides distal to the maxillary canines and canine retraction was carried out using NiTi closed coil springs (150gm) and direct anchorage with miniscrew implants. The second MOP was performed on experimental side one month after the first MOP. The rate of canine movement was assessed using 3D model superimposition over a period of six months. The type of tooth movement, anchorage loss and levels of IL-1ß were also evaluated. RESULTS: Sixteen patients (mean age, 17.87±3.34 years) were analysed for a rate of canine retraction, anchorage loss, and type of tooth movement, while 15 patients were analysed for IL-1ß. The rate was significantly higher on two-time MOP side after two months (P<0.001). No statistical difference was found in anchorage loss and controlled tipping of canines was observed. The IL-1ß levels immediately after 2nd MOP were significantly higher than 1st MOP (P<0.001). CONCLUSION: The two-time intervention of MOP is more efficacious than one-time MOP in accelerating tooth movement.


Subject(s)
Cuspid , Mouth , Adolescent , Adult , Face , Gingival Crevicular Fluid , Humans , Tooth Movement Techniques , Young Adult
3.
Anticancer Agents Med Chem ; 20(16): 1943-1955, 2020.
Article in English | MEDLINE | ID: mdl-32560618

ABSTRACT

BACKGROUND: The high mobility group box 1 (hmgb1) is one of the frequently over-expressed genes whose aberrant expression is reported in a number of human cancers. Various strategies are underway to inhibit hmgb1 expression in cancer cells having considerable therapeutic value. OBJECTIVE: The present work involves selective transcriptional inhibition of the hmgb1 gene using selective DNA triplex structure-based gene technology. Here, the promoter region of the hmgb1 gene at position (-183 to -165) from the transcription start site as a target was selected using bioinformatic tools. METHODS: The DNA triplex formation by the DNA of the target gene and TFO was confirmed using UV absorption spectroscopy, Circular Dichroism, and Isothermal Calorimetry. RESULTS: Treatment of HepG2 cell with specific Triplex-forming Oligonucleotide significantly downregulated HMGB1 expression level at mRNA and protein levels by 50%, while the classical anticancer drugs, actinomycin/ adriamycin as positive controls showed 65% and the combination of TFO and drug decreased by 70%. The anti-proliferative effects of TFO correlated well with the fact of accumulation of cells in the Go phase and apoptotic cell death. Further, the binding of anti-cancer drugs to hmgb1 is stronger in DNA triplex state as compared to hmgb1 alone, suggesting the combination therapy as a better option. CONCLUSION: Therefore, the ability of hmgb1 targeted triplex-forming oligonucleotide in combination with triplex selective anticancer drug holds promise in the treatment of malignancies associated with hmgb1 overexpression. The result obtained may open up new vistas to provide a basis for the rational drug design and searching for high-affinity ligands with a high triplex selectivity.


Subject(s)
Antineoplastic Agents/pharmacology , HMGB1 Protein/antagonists & inhibitors , Oligonucleotides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Computational Biology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HMGB1 Protein/genetics , Hep G2 Cells , Humans , Molecular Structure , Oligonucleotides/chemical synthesis , Oligonucleotides/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
4.
J Oral Biol Craniofac Res ; 10(2): 93-98, 2020.
Article in English | MEDLINE | ID: mdl-32211284

ABSTRACT

The peri-miniscrew implant crevicular fluid is analogous to gingival crevicular fluid, and its contents reflect the state of inflammation and health during the life of the miniscrews in the mouth. The stability of MSI is fundamental to its role as an anchorage. This study aimed to evaluate transforming growth factor-beta one (TGF-ß1) of the peri-miniscrew implant crevicular fluid (PMICF), on implant insertion, pre- and post-loading of MSIs to find a clue to their role in the stability of MSI. Fifty-two MSIs sites were placed in the mouths of 13 patients aged 12-26 years undergoing orthodontic treatment. PMICF was collected using micro-pipettes at T1 (day 0, 1 h after MSI implantation), T2 (day 1), T3/baseline (day 21, preloading of MSI), T4 (day 21, 1 h post loading), T5 (day 22, 1 day post loading), T6 (day 43, 3 weeks post loading). The levels of TGF-ß1 were estimated by enzyme-linked immunosorbent assay (ELISA). The data were subjected to statistical analysis. Of the 52 MSIs, 20 MSIs failed at T3. In the case of successful MSIs, the TGF-ß1 levels were found to monotonously decrease from T1 (~1400 pg/mL) until T3 (~700 pg/mL) and saturate thereafter. In the case of failed MSIs, the levels of TGF-ß1 at various time periods were approximately constant and of much lower value than corresponding time periods of successful MSIs. This study highlights the role of TGF- ß1 in bone metabolism around miniscrew reflecting the state of inflammation from 1 h post-implantation.

5.
Front Neurosci ; 13: 1054, 2019.
Article in English | MEDLINE | ID: mdl-31680804

ABSTRACT

Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by an expanded (GAA) trinucleotide repeat in the FXN gene. The extended repeats expansion results in reduced transcription and, thereby, decreased expression of the mitochondrial protein, frataxin. Given the ongoing drug trials, identification of reliable and easily accessible biomarkers for monitoring disease progression and therapeutic intervention is a foremost requirement. In this study, comparative proteomic profiling of PBMC proteins from FRDA patients and age- and gender-matched healthy controls was done using 2D-Differential in-Gel Electrophoresis (2D-DIGE). Protein-protein interaction (PPI) was analyzed using BioGRID and STRING pathway analysis tools. Using biological variance analysis (BVA) and LC/MS, we found eight differentially expressed proteins with fold change ≥1.5; p ≤ 0.05. Based on their cellular function, the identified proteins showed a strong pathological role in neuroinflammation, cardiomyopathy, compromised glucose metabolism, and iron transport, which are the major clinical manifestations of FRDA. Protein-protein network analysis of differentially expressed proteins with frataxin further supports their involvement in the pathophysiology of FRDA. Considering their crucial role in the cardiac and neurological complications, respectively, the two down-regulated proteins, actin α cardiac muscle 1 (ACTC1) and pyruvate dehydrogenase E1 component subunit ß (PDHE1), are suggested as potential prognostic markers for FRDA.

6.
Biometals ; 32(2): 307-315, 2019 04.
Article in English | MEDLINE | ID: mdl-30874991

ABSTRACT

Friedreich's ataxia (FRDA), a progressive neurodegenerative disorder caused by trinucleotide (GAA) repeat expansion in frataxin (fxn) gene which results in decreased levels of frataxin protein. Insufficient frataxin levels leads to iron and copper deposits in the brain and cardiac cells. A total of hundred and twenty patients, suspected of FRDA were screened for the (GAA) repeats in the fxn gene and only confirmed patients (n = 25) were recruited in the study. The total Iron and total copper concentrations were measured in blood plasma using Nitro PAPS and Dibrom PAESA method, respectively both in patients and age, sex matched healthy controls. The iron levels mean ± SD (6.2 ± 3.8) in plasma of FRDA patients were found to be significantly decreased as compared to healthy controls mean ± SD (15.2 ± 4.2). A similar trend was observed in case of plasma copper levels in FRDA patient (8.15 ± 4.6) as compared to controls (17.5 ± 3.40). Present results clearly prove abnormal distribution of extra-cellular iron in FRDA patients, which is in accordance with the well established fact of intracellular iron overload, which is the key feature of the pathogenesis of this disease. This can be of importance in understanding the pathophysiology of the disease in association with frataxin/iron. It appears that intracellular sequestration of trace metals in FRDA patients (due to low frataxin) results in their sub-optimal levels in blood plasma (extra-cellular) an observation that can find prognostic application in clinical trials.


Subject(s)
Copper/blood , Friedreich Ataxia/blood , Friedreich Ataxia/pathology , Iron/blood , Friedreich Ataxia/genetics , Humans , Trinucleotide Repeat Expansion/genetics
7.
Int J Biol Macromol ; 125: 1156-1167, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30579900

ABSTRACT

Survival of the Acinetobacter baumannii inside host requires different micronutrients such as iron, but their bioavailability is limited because of nutritional immunity created by host. A. baumannii has to develop mechanisms to acquire nutrient iron during infection. The present study is an attempt to identify membrane proteins involved in iron sequestration mechanism of A. baumannii using two-dimensional electrophoresis and LC-MS/MS analysis. The identified iron-regulated membrane protein (IRMP) of A. baumannii was used for its interaction studies with different siderophores, and designing of the inhibitor against A. baumannii targeting this IRMP. Membrane proteomic results identified over-expression of four membrane proteins (Fhu-E receptor, ferric-acinetobactin receptor, ferrienterochelin receptor, and ferric siderophore receptor) under iron-limited condition. A. baumannii produces siderophores that have good interaction with the FhuE receptor. Result also showed that FhuE receptor has interaction with siderophores produced by other bacteria. Interaction of FhuE receptor and siderophores helps in iron sequestration and survival of Acinetobacter under nutritional immunity imposed by the host. Hence it becomes essential to find a potential inhibitor for the FhuE receptor that can inhibit the survival of A. baumannii in the host. In-silico screening, and molecular mechanics studies identified ZINC03794794 and ZINC01530652 as a likely lead to design inhibitor against the FhuE receptor of A. baumannii. The designed inhibitor is experimentally validated for its antibacterial activity on the A. baumannii. Therefore, designed inhibitor interferes with the iron acquisition mechanism of Acinetobacter hence may prove useful for preventing infection caused by A. baumannii by limiting nutrient availability.


Subject(s)
Acinetobacter baumannii/drug effects , Acinetobacter baumannii/metabolism , Iron-Regulatory Proteins/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Proteomics , Siderophores/pharmacology , Carbapenems/chemistry , Carbapenems/pharmacology , Chromatography, Liquid , Iron/chemistry , Iron-Regulatory Proteins/chemistry , Membrane Proteins/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Proteome , Proteomics/methods , Siderophores/chemistry , Tandem Mass Spectrometry
9.
Neuropediatrics ; 49(1): 35-43, 2018 02.
Article in English | MEDLINE | ID: mdl-29179232

ABSTRACT

Friedreich's ataxia (FRDA) is a multisystem disease affecting the predominately nervous system, followed by muscle, heart, and pancreas. Current research focused on therapeutic interventions aimed at molecular amelioration, but there are no reliable noninvasive signatures available to understand disease pathogenesis. The present study investigates the alterations of plasma cell-free microRNAs (miRNAs) in FRDA patients and attempts to find the significance in relevance with the pathogenesis. Total RNA from the plasma of patients and healthy controls were subjected to miRNA microarray analysis using Agilent Technologies microarray platform. Differentially regulated miRNAs were validated by SYBR-green real-time polymerase chain reaction (Thermo Fisher Scientific). The study identified 20 deregulated miRNAs (false discovery rate < 0.01, fold change ≥ 2.0 ≤) in comparison with healthy controls; out of which 17 miRNAs were upregulated, and 3 miRNAs were downregulated. Target and pathway analysis of these miRNAs have shown association with neurodegenerative and other clinical features in FRDA. Further validation (n = 21) identified a set of significant (p < 0.05) deregulated miRNAs; hsa-miR-15a-5p, hsa-miR-26a-5p, hsa-miR-29a-3p, hsa-miR-223-3p, hsa-24-3p, and hsa-miR-21-5p in comparison with healthy controls. These miRNAs were reported to influence various pathological features associated with FRDA. The present study is expected to aid in the understanding of disease pathogenesis.


Subject(s)
Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Friedreich Ataxia , Adult , Case-Control Studies , Female , Friedreich Ataxia/blood , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Male , Microarray Analysis , Middle Aged , RNA, Messenger/metabolism , ROC Curve , Trinucleotide Repeats/genetics , Frataxin
12.
Anticancer Agents Med Chem ; 17(12): 1698-1709, 2017.
Article in English | MEDLINE | ID: mdl-28270074

ABSTRACT

BACKGROUND: HMGB1 (High Mobility Group Box-1) is a very versatile highly abundant architectural protein that plays multiple roles in human health and diseases. Under physiological condition it serves as an amazing assortment of roles in different compartments of cell. The reported high expression of HMGB1 in almost all types of human cancers and inflammatory diseases makes it a critical molecular therapeutic target. OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). METHOD: The interaction of actinomycin and adriamycin to 21R*R•Y DNA triplex was studied using UV melting profiles, CD spectroscopy, spectrofluorimetry and Isothermal titration calorimetry. The 21R*R•Y formation was confirmed from biphasic thermal melting profiles, continuous variation method, analysis of CD marker band and thermodynamic parameters. RESULTS: The binding of ADM and ACT to 21R*R•Y was characterized by hypochromic and bathochromic shift in their respective absorption spectrum, quenching (ADM) and enhanced fluorescence (ACT) of steady-state fluorescence intensity, perturbation in the circular dichroic spectrum and change in thermal melting temperatures. The ITC profile and Scatchard plot analysis indicate non-cooperative and higher binding affinity of these drugs to 21R*R•Y compared to their corresponding duplexes. CONCLUSION: Therefore, combining these chemotherapeutic drugs with triplex forming oligonucleotide may offer new diagnostic and therapeutic options in targeting a gene of interest more specifically with fewer side effects. This study shows that ACT and ADM effectively recognize 21R*R•Y triplex DNA formed on the hmgb1 promoter region.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/chemistry , Dactinomycin/pharmacology , Doxorubicin/pharmacology , HMGB1 Protein/genetics , Molecular Targeted Therapy , Promoter Regions, Genetic , Thermodynamics , Circular Dichroism , HMGB1 Protein/drug effects , Humans , Nucleic Acid Conformation , Nucleic Acid Denaturation , Spectrum Analysis/methods
13.
J Biomol Struct Dyn ; 35(4): 689-703, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26923360

ABSTRACT

High-mobility group A1 (HMGA1) is a non-histone chromosomal protein, which is known as 'architectural' transcription factor that facilitates the assembly of 'enhanceosome.' Because of its elevated expression in a number of human malignancies, with barely minimal levels in healthy adults, HMGA1 is considered as potential 'tumor marker.' Therefore, we looked at the inhibition of hmga1 using anti-gene strategy, as an attractive therapeutic approach. This was achieved by two triplex forming oligonucleotides (TFOs), TFO1 and TFO2 targeted to the promoter of hmga1 at positions, -284--304 and -2800--2826, respectively. The stability of two DNA triplexes was characterized using a variety of biophysical and thermodynamics techniques and was confirmed by gel retardation assay using γ-32P [ATP]. The efficacy of TFOs on HMGA1 expression was evaluated in HeLa cells using MTT assay, Flow cytometry, Western blot, and RT-PCR. Results revealed that DNA Triplex1 formed by TFO1 is more stable and stronger than the corresponding Triplex2. Although both TFOs downregulated hmga1 expression at mRNA and protein levels and caused apoptotic cell death in HeLa cell line, TFO1 demonstrated a greater effect at low concentration which corroborates well with the stability data. Thus, TFO-mediated inhibition of hmga1 expression can be a promising strategy for the development of novel anti-cancer therapeutics.


Subject(s)
Apoptosis/genetics , DNA/genetics , HMGA1a Protein/antagonists & inhibitors , Oligonucleotides/genetics , Promoter Regions, Genetic/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Adult , Base Sequence , Female , HMGA1a Protein/genetics , Humans , Tumor Cells, Cultured
14.
J Neurol Sci ; 365: 82-8, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27206881

ABSTRACT

Friedreich's ataxia (FRDA) is one of the most devastating childhood onset neurodegenerative disease affecting multiple organs in the course of progression. FRDA is associated with mitochondrial dysfunction due to deficit in a nuclear encoded mitochondrial protein, frataxin. Identification of disease-specific biomarker for monitoring the severity remains to be a challenging topic. This study was aimed to identify whether circulating cell-free nuclear DNA (nDNA) and mitochondrial DNA (mtDNA) in blood plasma can be a potential biomarker for FRDA. Clinical information was assessed using International Cooperative Ataxia Rating Scale and the disease was confirmed using Long-range PCR for GAA repeat expansion within the gene encoding frataxin. The frataxin expression was measured using Western blot. Plasma nDNA and mtDNA levels were quantified by Multiplex real-time PCR. The major observation is that the levels of nDNA found to be increased, whereas mtDNA levels were reduced significantly in the plasma of FRDA patients (n=21) as compared to healthy controls (n=21). Further, plasma mtDNA levels showed high sensitivity (90%) and specificity (76%) in distinguishing from healthy controls with optimal cutoff indicated at 4.1×10(5)GE/mL. Interestingly, a small group of follow-up patients (n=9) on intervention with, a nutrient supplement, omega-3 fatty acid (a known enhancer of mitochondrial metabolism) displayed a significant improvement in the levels of plasma mtDNA, supporting our hypothesis that plasma mtDNA can be a potential monitoring or prognosis biomarker for FRDA.


Subject(s)
DNA, Mitochondrial/blood , Friedreich Ataxia/blood , Friedreich Ataxia/genetics , Adolescent , Biomarkers/blood , Blotting, Western , Child , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Female , Follow-Up Studies , Friedreich Ataxia/diet therapy , Humans , Iron-Binding Proteins/blood , Iron-Binding Proteins/genetics , Male , Polymerase Chain Reaction , ROC Curve , Severity of Illness Index , Treatment Outcome , Trinucleotide Repeat Expansion , Young Adult , Frataxin
15.
Curr Protein Pept Sci ; 17(8): 762-775, 2016.
Article in English | MEDLINE | ID: mdl-26916160

ABSTRACT

The High mobility group box 1 (HMGB1) protein is an extremely versatile, highly conserved nuclear protein, with its unique intracellular and extracellular functions mediated by its relatively simple domain structure. Within the nucleus, HMGB1 binds to DNA minor groove in a nonspecific manner and causes bends in the double helix thus helps in recruiting a number of DNA binding protein and transcription factors, to facilitate transcription of various genes. HMGB1 also helps in DNA repair, chromatin remodeling, V (D) J recombination, and assembly of nucleosome on the chromatin. On contrary, under pathological conditions HMGB1 displays inflammatory response by interaction with specific cell surface receptors like RAGE, TLR-4, TLR9, and TLR2 and activates NF-kB downstream signaling pathways. The upregulation of HMGB1 is directly associated with the pathogenesis of cancer, sepsis, ischemia, hemorrhagic shock, anorexia, rheumatic disease, periodontal disease etc. Therefore, HMGB1 has been considered as a promising target in the treatment of various human diseases. The interest in HMGB1 is evident and reflected in the exponential increase in the recent publications, and therefore there is a need for an update on the understanding of the role of HMGB1 in pathogenesis and its potential application of HMGB1 as a therapeutic target in a number of human diseases.


Subject(s)
Chromatin Assembly and Disassembly , DNA/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Neoplasms/metabolism , DNA/genetics , Gene Expression Regulation , HMGB1 Protein/genetics , Humans , Inflammation/genetics , NF-kappa B/metabolism , Neoplasms/genetics , Protein Binding , Signal Transduction
16.
J Biomol Struct Dyn ; 34(11): 2518-35, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26599132

ABSTRACT

Certain plant-derived alkaloids and flavonoids have shown propitious cytotoxic acitvity against different types of cancer, having deoxyribose nucleic acid (DNA) as their main cellular target. Flavopiridol, a semi-synthetic derivative of rohitukine (a natural compound isolated from Dysoxylum binectariferum plant), has attained much attention owing to its anticancer potential against various haematological malignancies and solid tumours. This work focuses on investigating interaction between flavopiridol and DNA at molecular level in order to decipher its underlying mechanism of action, which is not well understood. To define direct influence of flavopiridol on the structural, conformational and thermodynamic aspects of DNA, various spectroscopic and calorimetric techniques have been used. ATR-FTIR and SERS spectral outcomes indicate a novel insight into groove-directed-intercalation of flavopiridol into DNA via direct binding with nitrogenous bases guanine (C6=O6) and thymine (C2=O2) in DNA groove together with slight external binding to its sugar-phosphate backbone. Circular dichroism spectral analysis of flavopiridol-DNA complexes suggests perturbation in native B-conformation of DNA and its transition into C-form, which may be localized up to a few base pairs of DNA. UV-visible spectroscopic results illustrate dual binding mode of flavopiridol when interacts with DNA having association constant, Ka = 1.18 × 10(4) M(-1). This suggests moderate type of interaction between flavopiridol and DNA. Further, UV melting analysis also supports spectroscopic outcomes. Thermodynamically, flavopiridol-DNA complexation is an enthalpy-driven exothermic process. These conclusions drawn from this study could be helpful in unveiling mechanism of cytoxicity induced by flavopiridol that can be further applied in the development of flavonoid-based new chemotherapeutics with more specificity and better efficacy.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Flavonoids/chemistry , Models, Molecular , Molecular Conformation , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Thermodynamics , Circular Dichroism , Nucleic Acid Conformation , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
17.
J Biomol Struct Dyn ; 34(4): 689-704, 2016.
Article in English | MEDLINE | ID: mdl-25990537

ABSTRACT

Purine repeat sequences present in a gene are unique as they have high propensity to form unusual DNA-triple helix structures. Friedreich's ataxia is the only human disease that is well known to be associated with DNA-triplexes formed by purine repeats. The purpose of this study was to recognize the expanded purine repeats (EPRs) in human genome and find their correlation with cancer pathogenesis. We developed "PuRepeatFinder.pl" algorithm to identify non-overlapping EPRs without pyrimidine interruptions in the human genome and customized for searching repeat lengths, n ≥ 200. A total of 1158 EPRs were identified in the genome which followed Wakeby distribution. Two hundred and ninety-six EPRs were found in geneic regions of 282 genes (EPR-genes). Gene clustering of EPR-genes was done based on their cellular function and a large number of EPR-genes were found to be enzymes/enzyme modulators. Meta-analysis of 282 EPR-genes identified only 63 EPR-genes in association with cancer, mostly in breast, lung, and blood cancers. Protein-protein interaction network analysis of all 282 EPR-genes identified proteins including those in cadherins and VEGF. The two observations, that EPRs can induce mutations under malignant conditions and that identification of some EPR-gene products in vital cell signaling-mediated pathways, together suggest the crucial role of EPRs in carcinogenesis. The new link between EPR-genes and their functionally interacting proteins throws a new dimension in the present understanding of cancer pathogenesis and can help in planning therapeutic strategies. Validation of present results using techniques like NGS is required to establish the role of the EPR genes in cancer pathology.


Subject(s)
Genetic Association Studies , Genome, Human , Neoplasms/genetics , Purines , Repetitive Sequences, Nucleic Acid , Algorithms , Chromosome Mapping , DNA , Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Introns , Models, Statistical , Multigene Family , Neoplasms/metabolism , Nucleic Acid Conformation , Protein Interaction Maps
18.
J Biomol Struct Dyn ; 34(4): 877-91, 2016.
Article in English | MEDLINE | ID: mdl-26084422

ABSTRACT

High mobility group A1 (HMGA1), a non-histone chromosomal protein, is highly expressed in a wide range of human cancers including cervical, breast, and prostate cancers. Therefore, hmga1 gene is considered as an attractive potential target for anticancer drugs. We have chosen 27 bp DNA sequence from a regulatory region of hmga1 promoter and studied its interaction with adriamycin (ADM) and in vitro expression of HMGA1 in the presence of ADM in HeLa cell line. A variety of biophysical techniques were employed to understand the characteristics of [DNA-ADM] complex. Spectrophotometric titration data, DNA denaturation profiles, and quenching of fluorescence of ADM in the presence of DNA demonstrated a strong complexation between DNA and ADM with a high binding affinity (Ka) of 1.3 × 10(6) M(-1) and a stoichiometry of 1:3 (drug:nucleotide). The energetics of binding obtained from isothermal titration calorimetry and differential scanning calorimetry suggest the binding to be exothermic and enthalpy (∆H, -6.7 ± 2.4 kcal M(-1)) and entropy (TΔS, 18.5 ± 6.4 kcal M(-1)) driven (20°C), which is typical of intercalative mode of binding. Further, results on decreased expression (by ~70%) of HMGA1 both at mRNA and protein levels in association with the observed cell death (by ~75%) in HeLa cell line, clearly confirm that ADM does target hmga1; however, the effect of ADM on genes other than hmga1 either directly or via hmga1-mediated pathways cannot be ruled out in the observed cytotoxicity. Therefore, hmga1 in general and particularly the regulatory region is a promising target for therapeutic strategy in combating cancer.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Doxorubicin/chemistry , HMGA Proteins/genetics , Regulatory Sequences, Nucleic Acid , Antibiotics, Antineoplastic/pharmacology , Calorimetry, Differential Scanning , Cell Death/genetics , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , HMGA Proteins/antagonists & inhibitors , Humans , Molecular Structure , Nucleic Acid Conformation , Protein Binding , Structure-Activity Relationship , Thermodynamics , Transition Temperature , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
19.
Data Brief ; 5: 218-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26543885

ABSTRACT

Purine repeats are randomly distributed in the human genome, however, they show potential role in the transcriptional deregulation of genes. Presence of long tracks of purine repeats in the genome can disturb its integrity and interfere with the cellular behavior by introducing mutations and/or triple stranded structure formation in DNA. Our data revealed interesting finding that a majority of genes carrying purine repeats, of length n≥200, were down regulated and found to be linked with several brain related diseases [1]. The unique feature of the purine repeats found in the present study clearly manifests their significant application in developing therapeutics for neurological diseases.

20.
Gene ; 572(2): 175-83, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26149656

ABSTRACT

Purine repeat sequences present in the human genome are known to act as hotspots for mutations leading to chromosomal imbalances. It is established that large purine repeats (PRs) form stable DNA triplex structure which can inhibit gene expression. Friedreich's ataxia (FRDA), the autosomal neurodegenerative disorder is the only human disease known so far, where a large purine (GAA) repeat in the FXN gene is known to inhibit the expression of frataxin protein. We explored the hidden purine repeats (PRn with n ≥ 200) if any, in the human genome to find out how they are associated with neurological disorders. The results showed 28 PRs, which are mostly restricted to the intronic regions. Interestingly, the transcriptome expression analysis of PR-carrying genes (PR-genes) revealed that most of them are down-regulated in neurological disorders (autism, Alzheimer's disease, schizophrenia, epilepsy, mental retardation, Parkinson's disease, brain tumor) as compared to that in healthy controls. The altered gene expression in brain disorders can be interpreted in terms of a possible expansion of purine repeats leading to formation of very stable DNA-triplex and/or alleviation of the repair enzymes and/or other unknown cellular factors. Interactome analysis identified four PR-genes in signaling pathways whose dysregulation is correlated directly with pathogenesis: GRK5 and KLK6 in Alzheimer's disease; FGF14 in craniosynostosis, mental retardation and FLT1 in neuroferritinopathy. By virtue of being mutational hotspots and their ability to form DNA-triplex, purine repeats in genome disturb the genome integrity and interfere with the transcriptional regulation. However, validation of the disease linkage of PR-genes can be validated using knock-out techniques.


Subject(s)
DNA/chemistry , Nervous System Diseases/genetics , Purines/metabolism , Trinucleotide Repeats , Computational Biology/methods , Down-Regulation , Fibroblast Growth Factors/genetics , G-Protein-Coupled Receptor Kinase 5/genetics , Gene Expression Regulation , Humans , Introns , Kallikreins/genetics , Models, Molecular , Vascular Endothelial Growth Factor Receptor-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...