Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Mol Biol Rep ; 51(1): 729, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862809

ABSTRACT

BACKGROUND: Preclinical and clinical studies over the past several decades have indicated the potential value of metformin, a widely utilized treatment for Type 2 diabetes, in prostate cancer therapy. Notably, these studies demonstrated metformin's pleiotropic effects on several molecular and metabolic pathways, such as androgen signaling, cell cycle, and cellular bioenergetics. In this study we investigated the role of metformin in regulating intracellular redox status and cell survival in LNCaP prostate cancer cells. METHODS AND RESULTS: The cytotoxic effects of metformin with or without the presence of SBI0206965 (AMPK inhibitor) on LNCaP cells were determined using MTT and trypan blue exclusion assays. Seahorse XP extracellular analysis, Liquid Chromatography/ Mass Spectrophotometry (LC/MS), and 2,7- and Dichlorofluoresin diacetate (DCFDA) assay were used to assess the effects of metformin on cellular bioenergetics, redox status, and redox-related metabolites. mRNA expression and protein concentration of redox-related enzymes were measured using Real Time-qPCR and ELISA assay, respectively. Independently of AMP-activated protein kinase, metformin exhibited a dose- and time-dependent inhibition of LNCaP cell survival, a response mitigated by glutathione or N-acetylcysteine (ROS scavengers) treatment. Notably, these findings were concomitant with a decline in ATP levels and the inhibition of oxidative phosphorylation. The results further indicated metformin's induction of reactive oxygen species, which significantly decreased glutathione levels and the ratio of reduced to oxidized glutathione, as well as the transsulfuration metabolite, cystathionine. Consistent with an induction of oxidative stress condition, metformin increased mRNA levels of the master redox transcription factor Nrf-2 (nuclear factor erythroid-derived 2-like), as well as transsulfuration enzymes cystathionine beta-synthase and cystathionase and GSH synthesis enzymes γ-glutamylcysteine synthetase and glutathione synthetase. CONCLUSION: Our findings highlight multiple mechanisms by which metformin-induced formation of reactive oxygen species may contribute to its efficacy in prostate cancer treatment, including promotion of oxidative stress, Nrf2 activation, and modulation of redox-related pathways, leading to its anti-survival action.


Subject(s)
Cell Survival , Metformin , Oxidative Stress , Prostatic Neoplasms , Reactive Oxygen Species , Metformin/pharmacology , Humans , Male , Oxidative Stress/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Cell Survival/drug effects , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Oxidation-Reduction/drug effects , Glutathione/metabolism , AMP-Activated Protein Kinases/metabolism , Energy Metabolism/drug effects
2.
Mol Ther Oncolytics ; 25: 57-68, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35399604

ABSTRACT

Most advanced-stage ovarian cancer patients, including those with epithelial ovarian cancer (EOC), develop recurrent disease and acquisition of resistance to chemotherapy, leading to limited treatment options. Decrease in Let7b miRNA levels in clinical ovarian cancer has been associated with chemoresistance, increased proliferation, invasion, and relapse in EOC. We have established a murine EOC relapsed model by administering paclitaxel (PTX) and stopping therapy to allow for tumor regrowth. Global microRNA profiling in the relapsed tumor showed significant downregulation of Let7b relative to untreated tumors. Here, we report the use of hyaluronic acid (HA)-based nanoparticle formulation that can deliver Let7b miRNA mimic to tumor cells and achieve cellular programming both in vitro and in vivo. We demonstrate that a therapeutic combination of Let7b miRNA and PTX leads to significant improvement in anti-tumor efficacy in the relapsed model of EOC. We further demonstrate that the combination therapy is safe for repeated administration. This novel approach of cellular reprogramming of tumor cells using clinically relevant miRNA mimic in combination with chemotherapy could enable more effective therapeutic outcomes for patients with advanced-stage relapsed EOC.

3.
Transl Psychiatry ; 9(1): 295, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719518

ABSTRACT

D4 dopamine receptor (D4R) activation uniquely promotes methylation of plasma membrane phospholipids, utilizing folate-derived methyl groups provided by methionine synthase (MS). We evaluated the impact of D4R expression on folate-dependent phospholipid methylation (PLM) and MS activity, as well as cellular redox and methylation status, in transfected CHO cells expressing human D4R variants containing 2, 4, or 7 exon III repeats (D4.2R, D4.4R, D4.7R). Dopamine had no effect in non-transfected CHO cells, but increased PLM to a similar extent for both D4.2R- and D4.4R-expressing cells, while the maximal increase was for D4.7R was significantly lower. D4R expression in CHO cells decreased basal MS activity for all receptor subtypes and conferred dopamine-sensitive MS activity, which was greater with a higher number of repeats. Consistent with decreased MS activity, D4R expression decreased basal levels of methylation cycle intermediates methionine, S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH), as well as cysteine and glutathione (GSH). Conversely, dopamine stimulation increased GSH, SAM, and the SAM/SAH ratio, which was associated with a more than 2-fold increase in global DNA methylation. Our findings illustrate a profound influence of D4R expression and activation on MS activity, coupled with the ability of dopamine to modulate cellular redox and methylation status. These previously unrecognized signaling activities of the D4R provide a unique link between neurotransmission and metabolism.


Subject(s)
DNA Methylation , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Receptors, Dopamine D4/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Methionine/metabolism , Phospholipids/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
4.
DNA Cell Biol ; 38(6): 561-571, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30920300

ABSTRACT

Gulf War Illness (GWI) affects about 25% of Persian Gulf veterans with a cluster of chronic symptoms, including immune dysfunction and neurological issues. Recent studies implicate gene expression changes in immune function to be associated with GWI. Since DNA methylation can regulate such changes in gene expression, and disruption of DNA methylation pattern is implicated in various immune and neurological diseases, we aimed to study the DNA methylation patterns in peripheral blood mononuclear cells from GWI patients. Global DNA methylation levels were similar in GWI patients and controls. However, the genome-wide microarray technology detected 10,767 differentially methylated CpG sites across gene regulatory elements and within coding regions. Approximately 88% of them were hypermethylated in GWI patients. The separate analysis found 776 differentially methylated gene promoters (DMP), which were predominantly hypermethylated. Pyrosequencing validation confirmed microarray results. Functional analysis revealed that majority of the DMPs belonged to genes responsible for metabolism and immune system. This is the first pilot human study characterizing genome-wide epigenetic changes associated with GWI. It suggests a significant contribution of epigenetic dysfunction in GWI. Moreover, it supports the dysregulation of immune function in GWI. Lastly, it suggests studies with the larger cohort to validate our findings.


Subject(s)
DNA Methylation , Persian Gulf Syndrome/genetics , Adult , Cohort Studies , CpG Islands , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Persian Gulf Syndrome/immunology , Pilot Projects , Promoter Regions, Genetic , Sequence Analysis, DNA
5.
PLoS One ; 13(7): e0201066, 2018.
Article in English | MEDLINE | ID: mdl-30036399

ABSTRACT

BACKGROUND: Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex condition involving multiple organ systems and characterized by persistent/relapsing debilitating fatigue, immune dysfunction, neurological problems, and other symptoms not curable for at least 6 months. Disruption of DNA methylation patterns has been tied to various immune and neurological diseases; however, its status in ME/CFS remains uncertain. Our study aimed at identifying changes in the DNA methylation patterns that associate with ME/CFS. METHODS: We extracted genomic DNA from peripheral blood mononuclear cells from 13 ME/CFS study subjects and 12 healthy controls and measured global DNA methylation by ELISA-like method and site-specific methylation status using Illumina MethylationEPIC microarrays. Pyrosequencing validation included 33 ME/CFS cases and 31 controls from two geographically distant cohorts. RESULTS: Global DNA methylation levels of ME/CFS cases were similar to those of controls. However, microarray-based approach allowed detection of 17,296 differentially methylated CpG sites in 6,368 genes across regulatory elements and within coding regions of genes. Analysis of DNA methylation in promoter regions revealed 307 differentially methylated promoters. Ingenuity pathway analysis indicated that genes associated with differentially methylated promoters participated in at least 15 different pathways mostly related to cell signaling with a strong immune component. CONCLUSIONS: This is the first study that has explored genome-wide epigenetic changes associated with ME/CFS using the advanced Illumina MethylationEPIC microarrays covering about 850,000 CpG sites in two geographically distant cohorts of ME/CFS cases and matched controls. Our results are aligned with previous studies that indicate a dysregulation of the immune system in ME/CFS. They also suggest a potential role of epigenetic de-regulation in the pathobiology of ME/CFS. We propose screening of larger cohorts of ME/CFS cases to determine the external validity of these epigenetic changes in order to implement them as possible diagnostic markers in clinical setting.


Subject(s)
DNA Methylation , Fatigue Syndrome, Chronic/metabolism , Cohort Studies , CpG Islands , Epigenesis, Genetic , Fatigue Syndrome, Chronic/genetics , Female , Humans , Microarray Analysis , Middle Aged , Promoter Regions, Genetic
6.
PLoS One ; 12(7): e0181978, 2017.
Article in English | MEDLINE | ID: mdl-28738082

ABSTRACT

Sleep is critical for repair as well as the rejuvenation processes in the body and many of these functions are regulated via underlying cellular metabolic homeostasis. Changes in sleep pattern are reported to alter such metabolic function resulting in altered disease susceptibility or behavior. Here, we measured the extent to which overnight total sleep deprivation (SD) in young adult humans can influence systemic (plasma-derived) redox-metabolism including the major antioxidant, glutathione as well as DNA methylation levels. Nineteen participants (n = 19, µ age = 21, SD = 3.09) underwent morning testing before and after overnight total SD. Biochemical measures before and after SD revealed that glutathione, ATP, cysteine, and homocysteine levels were significantly reduced following one night of sleep deprivation (all p's < 0.01). Parallel to the well-recognized fact that sleep deprivation (maintaining wakefulness) uses up metabolic reserves, we observed that morning cortisol levels were blunted after sleep deprivation. There were no significant correlations between self-reported or actigraphy-measured sleep and the biochemical measurements, strongly indicating that prior sleep behavior did not have any direct influence on the biochemical measures taken at baseline or after sleep deprivation. Results from the current investigation supports the previous literature implicating the induction of oxidative stress and ATP depletion with sleep deprivation. Furthermore, such altered antioxidant status can also induce downstream epigenetic changes. Although we did not measure the specific genes that were altered under the influence of such sleep deprivation, such epigenetic changes could potentially contribute towards disease predisposition.


Subject(s)
Epigenesis, Genetic/genetics , Sleep Deprivation/genetics , Sleep Deprivation/metabolism , Sleep/genetics , Sleep/physiology , Actigraphy/methods , Adult , Circadian Rhythm/genetics , Circadian Rhythm/physiology , DNA Methylation/genetics , Epigenomics/methods , Female , Humans , Male , Oxidation-Reduction , Oxidative Stress/genetics , Sleep Deprivation/physiopathology , Wakefulness/genetics , Wakefulness/physiology , Young Adult
7.
PLoS One ; 11(1): e0146797, 2016.
Article in English | MEDLINE | ID: mdl-26799654

ABSTRACT

Many studies indicate a crucial role for the vitamin B12 and folate-dependent enzyme methionine synthase (MS) in brain development and function, but vitamin B12 status in the brain across the lifespan has not been previously investigated. Vitamin B12 (cobalamin, Cbl) exists in multiple forms, including methylcobalamin (MeCbl) and adenosylcobalamin (AdoCbl), serving as cofactors for MS and methylmalonylCoA mutase, respectively. We measured levels of five Cbl species in postmortem human frontal cortex of 43 control subjects, from 19 weeks of fetal development through 80 years of age, and 12 autistic and 9 schizophrenic subjects. Total Cbl was significantly lower in older control subjects (> 60 yrs of age), primarily reflecting a >10-fold age-dependent decline in the level of MeCbl. Levels of inactive cyanocobalamin (CNCbl) were remarkably higher in fetal brain samples. In both autistic and schizophrenic subjects MeCbl and AdoCbl levels were more than 3-fold lower than age-matched controls. In autistic subjects lower MeCbl was associated with decreased MS activity and elevated levels of its substrate homocysteine (HCY). Low levels of the antioxidant glutathione (GSH) have been linked to both autism and schizophrenia, and both total Cbl and MeCbl levels were decreased in glutamate-cysteine ligase modulatory subunit knockout (GCLM-KO) mice, which exhibit low GSH levels. Thus our findings reveal a previously unrecognized decrease in brain vitamin B12 status across the lifespan that may reflect an adaptation to increasing antioxidant demand, while accelerated deficits due to GSH deficiency may contribute to neurodevelopmental and neuropsychiatric disorders.


Subject(s)
Aging/metabolism , Autistic Disorder/metabolism , DNA Methylation/genetics , Frontal Lobe/metabolism , Gene Expression Regulation/genetics , Schizophrenia/metabolism , Vitamin B 12/analogs & derivatives , Vitamin B 12/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antioxidants/metabolism , Autistic Disorder/genetics , Child , Child, Preschool , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Humans , Infant , Infant, Newborn , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Schizophrenia/genetics , Vitamin B 12/genetics , Young Adult
8.
Nutr Metab (Lond) ; 12: 54, 2015.
Article in English | MEDLINE | ID: mdl-26664459

ABSTRACT

BACKGROUND: Casein-free, gluten-free diets have been reported to mitigate some of the inflammatory gastrointestinal and behavioral traits associated with autism, but the mechanism for this palliative effect has not been elucidated. We recently showed that the opioid peptide beta-casomorphin-7, derived from bovine (bBCM7) milk, decreases cysteine uptake, lowers levels of the antioxidant glutathione (GSH) and decreases the methyl donor S-adenosylmethionine (SAM) in both Caco-2 human GI epithelial cells and SH-SY5Y human neuroblastoma cells. While human breast milk can also release a similar peptide (hBCM-7), the bBCM7 and hBCM-7 vary greatly in potency; as the bBCM-7 is highly potent and similar to morphine in it's effects. Since SAM is required for DNA methylation, we wanted to further investigate the epigenetic effects of these food-derived opioid peptides. In the current study the main objective was to characterize functional pathways and key genes responding to DNA methylation effects of food-derived opioid peptides. METHODS: SH-SY5Y neuroblastoma cells were treated with 1 µM hBCM7 and bBCM7 and RNA and DNA were isolated after 4 h with or without treatment. Transcriptional changes were assessed using a microarray approach and CpG methylation status was analyzed at 450,000 CpG sites. Functional implications from both endpoints were evaluated via Ingenuity Pathway Analysis 4.0 and KEGG pathway analysis was performed to identify biological interactions between transcripts that were significantly altered at DNA methylation or transcriptional levels (p < 0.05, FDR <0.1). RESULTS: Here we show that hBCM7 and bBCM7, as well as morphine, cause epigenetic changes affecting gene pathways related to gastrointestinal disease and inflammation. These epigenetic consequences exhibited the same potency order as opiate inhibition of cysteine uptake insofar as hBCM7 was less potent than bBCM7, which was less potent than morphine. CONCLUSION: Our findings indicate that epigenetic effects of milk-derived opiate peptides may contribute to GI dysfunction and inflammation in sensitive individuals. While the current study was performed using SH-SY5Y neuronal cellular models, similar actions on other cells types might combine to cause symptoms of intolerance. These actions may provide a potential contributing mechanism for the beneficial effects of a casein-free diet in alleviating gastrointestinal symptoms in neurological conditions including autism and other conditions. Lastly, our study also contributes to the evolving awareness of a "gut-brain connection".

9.
Mol Cancer Ther ; 14(4): 1075-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25657335

ABSTRACT

Ovarian cancer is a dreadful disease estimated to be the second most common gynecologic malignancy worldwide. Its current therapy, based on cytoreductive surgery followed by the combination of platinum and taxanes, is frequently complicated by the onset of multidrug resistance (MDR). The discovery that survivin, a small antiapoptotic protein, is involved in chemoresistance provided a new prospect to overcome MDR in cancer, because siRNA could be used to inhibit the expression of survivin in cancer cells. With this in mind, we have developed self-assembly polymeric micelles (PM) able to efficiently co-load an anti-survivin siRNA and a chemotherapeutic agent, such as paclitaxel (PXL; survivin siRNA/PXL PM). Previously, we have successfully demonstrated that the downregulation of survivin by using siRNA-containing PM strongly sensitizes different cancer cells to paclitaxel. Here, we have evaluated the applicability of the developed multifunctional PM in vivo. Changes in survivin expression, therapeutic efficacy, and biologic effects of the nanopreparation were investigated in an animal model of paclitaxel-resistant ovarian cancer. The results obtained in mice xenografed with SKOV3-tr revealed a significant downregulation of survivin expression in tumor tissues together with a potent anticancer activity of survivin siRNA/PXL PM, while the tumors remained unaffected with the same quantity of free paclitaxel. These promising results introduce a novel type of nontoxic and easy-to-obtain nanodevice for the combined therapy of siRNA and anticancer agents in the treatment of chemoresistant tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Delivery Systems , Drug Resistance, Neoplasm/genetics , Inhibitor of Apoptosis Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins/metabolism , Mice , Micelles , Microtubules/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , Survivin , Xenograft Model Antitumor Assays
10.
J Nutr Biochem ; 25(10): 1011-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25018147

ABSTRACT

Dietary interventions like gluten-free and casein-free diets have been reported to improve intestinal, autoimmune and neurological symptoms in patients with a variety of conditions; however, the underlying mechanism of benefit for such diets remains unclear. Epigenetic programming, including CpG methylation and histone modifications, occurring during early postnatal development can influence the risk of disease in later life, and such programming may be modulated by nutritional factors such as milk and wheat, especially during the transition from a solely milk-based diet to one that includes other forms of nutrition. The hydrolytic digestion of casein (a major milk protein) and gliadin (a wheat-derived protein) releases peptides with opioid activity, and in the present study, we demonstrate that these food-derived proline-rich opioid peptides modulate cysteine uptake in cultured human neuronal and gastrointestinal (GI) epithelial cells via activation of opioid receptors. Decreases in cysteine uptake were associated with changes in the intracellular antioxidant glutathione and the methyl donor S-adenosylmethionine. Bovine and human casein-derived opioid peptides increased genome-wide DNA methylation in the transcription start site region with a potency order similar to their inhibition of cysteine uptake. Altered expression of genes involved in redox and methylation homeostasis was also observed. These results illustrate the potential of milk- and wheat-derived peptides to exert antioxidant and epigenetic changes that may be particularly important during the postnatal transition from placental to GI nutrition. Differences between peptides derived from human and bovine milk may contribute to developmental differences between breastfed and formula-fed infants. Restricted antioxidant capacity, caused by wheat- and milk-derived opioid peptides, may predispose susceptible individuals to inflammation and systemic oxidation, partly explaining the benefits of gluten-free or casein-free diets.


Subject(s)
Antioxidants/pharmacology , Cysteine/metabolism , Epigenesis, Genetic , Opioid Peptides/pharmacology , Animals , Caco-2 Cells , Caseins/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gliadin/metabolism , Glutathione/metabolism , Humans , Hydrolysis , Intestinal Mucosa/metabolism , Linear Models , Milk/chemistry , Opioid Peptides/isolation & purification , Oxidation-Reduction , S-Adenosylmethionine/metabolism , Triticum/chemistry
11.
Front Neurosci ; 8: 444, 2014.
Article in English | MEDLINE | ID: mdl-25657617

ABSTRACT

Alcohol and other drugs of abuse, including psychostimulants and opioids, can induce epigenetic changes: a contributing factor for drug addiction, tolerance, and associated withdrawal symptoms. DNA methylation is a major epigenetic mechanism and it is one of more than 200 methylation reactions supported by methyl donor S-adenosylmethionine (SAM). Levels of SAM are controlled by cellular redox status via the folate and vitamin B12-dependent enzyme methionine synthase (MS). For example, under oxidative conditions MS is inhibited, diverting its substrate homocysteine (HCY) to the trans sulfuration pathway. Alcohol, dopamine, and morphine, can alter intracellular levels of glutathione (GSH)-based cellular redox status, subsequently affecting SAM levels and DNA methylation status. Here, existing evidence is presented in a coherent manner to propose a novel hypothesis implicating the involvement of redox-based epigenetic changes in drug addiction. Further, we discuss how a "gene priming" phenomenon can contribute to the maintenance of redox and methylation status homeostasis under various stimuli including drugs of abuse. Additionally, a new mechanistic rationale for the use of metabolic interventions/redox-replenishers as symptomatic treatment of alcohol and other drug addiction and associated withdrawal symptoms is also provided. Hence, the current review article strengthens the hypothesis that neuronal metabolism has a critical bidirectional coupling with epigenetic changes in drug addiction exemplified by the link between redox-based metabolic changes and resultant epigenetic consequences under the effect of drugs of abuse.

12.
PLoS One ; 8(2): e56927, 2013.
Article in English | MEDLINE | ID: mdl-23437274

ABSTRACT

The folate and vitamin B12-dependent enzyme methionine synthase (MS) is highly sensitive to cellular oxidative status, and lower MS activity increases production of the antioxidant glutathione, while simultaneously decreasing more than 200 methylation reactions, broadly affecting metabolic activity. MS mRNA levels in postmortem human cortex from subjects across the lifespan were measured and a dramatic progressive biphasic decrease of more than 400-fold from 28 weeks of gestation to 84 years was observed. Further analysis revealed alternative splicing of MS mRNA, including deletion of folate-binding domain exons and age-dependent deletion of exons from the cap domain, which protects vitamin B12 (cobalamin) from oxidation. Although three species of MS were evident at the protein level, corresponding to full-length and alternatively spliced mRNA transcripts, decreasing mRNA levels across the lifespan were not associated with significant changes in MS protein or methionine levels. MS mRNA levels were significantly lower in autistic subjects, especially at younger ages, and this decrease was replicated in cultured human neuronal cells by treatment with TNF-α, whose CSF levels are elevated in autism. These novel findings suggest that rather than serving as a housekeeping enzyme, MS has a broad and dynamic role in coordinating metabolism in the brain during development and aging. Factors adversely affecting MS activity, such as oxidative stress, can be a source of risk for neurological disorders across the lifespan via their impact on methylation reactions, including epigenetic regulation of gene expression.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Alternative Splicing , Autistic Disorder/genetics , Cerebral Cortex/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , Adolescent , Adult , Age Factors , Child , Child, Preschool , Female , Gene Expression Regulation , Gene Order , Humans , Infant , Infant, Newborn , Male , Models, Biological , Oxidation-Reduction , Protein Interaction Domains and Motifs/genetics , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sulfur/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vitamin B 12/metabolism , Young Adult
13.
Int J Pharm ; 441(1-2): 82-91, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23262426

ABSTRACT

The completion of human genome project has increased our knowledge of the molecular mechanisms of many diseases, including cancer, thus providing new opportunities for gene therapy. Antisense oligodeoxynucleotides (AsODN) possess great potential as sequence-specific therapeutic agents, which in contrast to classic treatments provide more efficient and target-specific approach to modulate disease-related genes. To be therapeutically effective, sufficient concentrations of intact AsODN must bypass membrane barriers and access the site of action. In this study, a dendrosome delivery strategy was designed to improve the encapsulation of AsODN in non-cationic liposomes to target PKC-α in lung cancer cells in vitro. Subcellular trafficking of fluorescently labeled AsODN was visualized using confocal microscopy. Uptake and expression of mRNA and target protein after AsODN delivery was measured by flow cytometry, qRT-PCR and Western blot analysis, respectively. Dendrosomes showed favorable physicochemical parameters: high encapsulation efficiency and uptake in serum-containing medium with no apparent cytotoxicity. AsODN encapsulated in dendrosome efficiently and specifically suppress the target gene at both mRNA and protein levels. Additional in vivo studies on the application of dendrosome as a delivery system for nucleic acid molecules may lead to improvement of this technology and facilitate the development of therapeutic antisense techniques.


Subject(s)
Down-Regulation , Lung Neoplasms/genetics , Oligodeoxyribonucleotides, Antisense/administration & dosage , Protein Kinase C-alpha/genetics , Blotting, Western , Cell Line, Tumor , Dendrimers/administration & dosage , Flow Cytometry , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Liposomes , Lung Neoplasms/therapy , Microscopy, Confocal , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Front Neurosci ; 6: 92, 2012.
Article in English | MEDLINE | ID: mdl-22740813

ABSTRACT

Homeostatic synaptic scaling in response to neuronal stimulus or activation, and due to changes in cellular niche, is an important phenomenon for memory consolidation, retrieval, and other similar cognitive functions (Turrigiano and Nelson, 2004). Neurological disorders and cognitive disabilities in autism, Rett syndrome, schizophrenia, dementia, etc., are strongly correlated to alterations in protein expression (both synaptic and cytoplasmic; Cajigas et al., 2010). This correlation suggests that efficient temporal regulation of synaptic protein expression is important for synaptic plasticity. In addition, equilibrium between mRNA processing, protein translation, and protein turnover is a critical sensor/trigger for recording synaptic information, normal cognition, and behavior (Cajigas et al., 2010). Thus a regulatory switch, which controls the lifespan, maturation, and processing of mRNA, might influence cognition and adaptive behavior. Here, we propose a two part novel hypothesis that methylation might act as this suggested coordinating switch to critically regulate mRNA maturation at (1) the pre-transcription level, by regulating precursor-RNA processing into mRNA, via other non-coding RNAs and their influence on splicing phenomenon, and (2) the post-transcription level by modulating the regulatory functions of ribonucleoproteins and RNA binding proteins in mRNA translation, dendritic translocation as well as protein synthesis and synaptic turnover. DNA methylation changes are well recognized and highly correlated to gene expression levels as well as, learning and memory; however, RNA methylation changes are recently characterized and yet their functional implications are not established. This review article provides some insight on the intriguing consequences of changes in methylation levels on mRNA life-cycle. We also suggest that, since methylation is under the control of glutathione anti-oxidant levels (Lertratanangkoon et al., 1997), the redox status of neurons might be the central regulatory switch for methylation-based changes in mRNA processing, protein expression, and turnover. Lastly, we also describe experimental methods and techniques which might help researchers to evaluate the suggested hypothesis.

15.
Nutrition ; 28(7-8): e27-32, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22541054

ABSTRACT

OBJECTIVE: To evaluate the association between suboptimal breast-feeding practices and autism spectrum disorders (ASDs). METHODS: A case-control study was conducted in 102 ASD cases and 102 matched healthy controls. RESULTS: Based on adjusted odds ratios from logistic regression models, ASD was found to be associated with the late initiation of breast-feeding (odds ratio 1.48, 95% confidence interval 1.01-3.1), a non-intake of colostrum (odds ratio 1.7, 95% confidence interval 1.03-4.3), prelacteal feeding, and bottle-feeding. The risk of ASD was found to decrease in a dose-response fashion over increasing periods of exclusive breast-feeding (P for trend = 0.04) and continued breast-feeding (P for trend = 0.001). CONCLUSION: The study indicates that increased ASD risk is generally associated with suboptimal breast-feeding practices.


Subject(s)
Breast Feeding , Child Development Disorders, Pervasive/epidemiology , Child Development , Adolescent , Breast Feeding/ethnology , Case-Control Studies , Child , Child Development Disorders, Pervasive/ethnology , Child Health Services , Child, Preschool , Colostrum , Diagnostic and Statistical Manual of Mental Disorders , Female , Hospitals, University , Humans , Logistic Models , Male , Oman/epidemiology , Outpatient Clinics, Hospital , Pilot Projects , Risk , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...