Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Anim Biotechnol ; 34(8): 3564-3577, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36811467

ABSTRACT

In countries where farming is largely subsistence, no pedigree records of farm animals are maintained at farmers' herd and scientific mating plans are not observed which leads to the accumulation of inbreeding and loss of production potential. Microsatellites have been widely used as reliable molecular markers to measure inbreeding. We attempted to correlate autozygosity estimated from microsatellite data with the inbreeding coefficient (F) calculated from pedigree data in Vrindavani crossbred cattle developed in India. The inbreeding coefficient was calculated from the pedigree of ninety-six Vrindavani cattle. Animals were further grouped into three groups viz. acceptable/low (F: 0-5%), moderate (F: 5-10%) and high (F: ≥10%), based on their inbreeding coefficients. The overall mean of the inbreeding coefficient was found to be 0.070 ± 0.007. A panel of twenty-five bovine-specific loci were chosen for the study according to ISAG/FAO. The mean FIS, FST, and FIT values were 0.0548 ± 0.025, 0.012 ± 0.001 and 0.0417 ± 0.025, respectively. There was no significant correlation between the FIS values obtained and the pedigree F values. The locus-wise individual autozygosity was estimated using the method-of-moments estimator (MME) formula for locus-specific autozygosity. The autozygosities ascribing to CSSM66 and TGLA53 were found to be significantly (p < .01 and p < .05, respectively) correlated with pedigree F values.


Subject(s)
Inbreeding , Polymorphism, Single Nucleotide , Cattle/genetics , Animals , Pedigree , Microsatellite Repeats/genetics , Reproduction
2.
Anesthesiology ; 137(2): 212-231, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35504002

ABSTRACT

BACKGROUND: Inhalational anesthetics are known to disrupt PDZ2 domain-mediated protein-protein interactions of the postsynaptic density (PSD)-95 protein. The aim of this study is to investigate the underlying mechanisms in response to early isoflurane exposure on synaptic PSD-95 PDZ2 domain disruption that altered spine densities and cognitive function. The authors hypothesized that activation of protein kinase-G by the components of nitric oxide (NO) signaling pathway constitutes a mechanism that prevents loss of early dendritic spines and synapse in neurons and cognitive impairment in mice in response to disruption of PDZ2 domain of the PSD-95 protein. METHODS: Postnatal day 7 mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active PSD-95 wild-type PDZ2 peptide or soluble guanylyl cyclase activator YC-1 along with their respective controls. Primary neurons at 7 days in vitro were exposed to isoflurane or PSD-95 wild-type PDZ2 peptide for 4 h. Coimmunoprecipitation, spine density, synapses, cyclic guanosine monophosphate-dependent protein kinase activity, and novel object recognition memory were assessed. RESULTS: Exposure of isoflurane or PSD-95 wild-type PDZ2 peptide relative to controls causes the following. First, there is a decrease in PSD-95 coimmunoprecipitate relative to N-methyl-d-aspartate receptor subunits NR2A and NR2B precipitate (mean ± SD [in percentage of control]: isoflurane, 54.73 ± 16.52, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 51.32 ± 12.93, P = 0.001). Second, there is a loss in spine density (mean ± SD [spine density per 10 µm]: control, 5.28 ± 0.56 vs. isoflurane, 2.23 ± 0.67, P < 0.0001; and PSD-95 mutant PDZ2 peptide, 4.74 ± 0.94 vs. PSD-95 wild-type PDZ2 peptide, 1.47 ± 0.87, P < 0.001) and a decrease in synaptic puncta (mean ± SD [in percentage of control]: isoflurane, 41.1 ± 14.38, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 50.49 ± 14.31, P < 0.001). NO donor or cyclic guanosine monophosphate analog prevents the spines and synapse loss and decline in the cyclic guanosine monophosphate-dependent protein kinase activity, but this prevention was blocked by soluble guanylyl cyclase or protein kinase-G inhibitors in primary neurons. Third, there were deficits in object recognition at 5 weeks (mean ± SD [recognition index]: male, control, 64.08 ± 10.57 vs. isoflurane, 48.49 ± 13.41, P = 0.001, n = 60; and female, control, 67.13 ± 11.17 vs. isoflurane, 53.76 ± 6.64, P = 0.003, n = 58). Isoflurane-induced impairment in recognition memory was preventable by the introduction of YC-1. CONCLUSIONS: Activation of soluble guanylyl cyclase or protein kinase-G prevents isoflurane or PSD-95 wild-type PDZ2 peptide-induced loss of dendritic spines and synapse. Prevention of recognition memory with YC-1, a NO-independent activator of guanylyl cyclase, supports a role for the soluble guanylyl cyclase mediated protein kinase-G signaling in countering the effects of isoflurane-induced cognitive impairment.


Subject(s)
Cognitive Dysfunction , Cyclic GMP-Dependent Protein Kinases , Disks Large Homolog 4 Protein , Isoflurane , Animals , Cognitive Dysfunction/chemically induced , Cyclic GMP-Dependent Protein Kinases/metabolism , Disks Large Homolog 4 Protein/metabolism , Female , Guanosine Monophosphate , Isoflurane/toxicity , Male , Mice , Nitric Oxide/metabolism , Peptides , Post-Synaptic Density , Signal Transduction , Soluble Guanylyl Cyclase , Synapses
3.
J Pediatr ; 227: 281-287, 2020 12.
Article in English | MEDLINE | ID: mdl-32599033

ABSTRACT

OBJECTIVE: To determine the average reported consent rate for published pediatric randomized controlled trials (RCTs) and whether this rate varies by trial characteristics. STUDY DESIGN: A review of pediatric RCTs published in Medline in 2009, 2010, or 2015 was performed. Secondary analyses of prior trials, trials including adults, trials not requiring consent, or trials with missing or unclear consent data were excluded. Consent rate was defined as the number of patients enrolled divided by number of eligible patients where families were approached. Random effects meta-regression was conducted to determine the weighted average consent rate. RESULTS: Of 2347 trials identified, 1651 were excluded. An additional 418 of 696 (60%) were excluded because the consent rate was missing or unclear. The average consent rate for 278 included RCTs was 82.6% (95% CI, 80.3%-84.8%) and was higher for vaccination compared with behavioral trials and for industry-funded compared with National Institutes of Health-funded or other government-funded trials. The average consent rate was <70% for 26% of included trials. Of these trials, US trials (28/77 [36.4%]) had a higher probability of a consent rate of <70% than non-US studies (35/64 [21.3%]) and multinational (9/37 [24.3%]) studies. There was slight variation by funding category. CONCLUSIONS: Although the average consent rate for published trials was reasonably high, approximately one-quarter of trials had consent rates of <70%. Consent rates reporting has improved over time, but remains suboptimal. Our findings should assist with the planning of future pediatric RCTs, although consent data from unpublished trials are also needed.


Subject(s)
Parental Consent/statistics & numerical data , Publishing/statistics & numerical data , Randomized Controlled Trials as Topic , Child , Humans , Pediatrics
4.
J Biol Chem ; 292(47): 19423-19440, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28982980

ABSTRACT

The widely used carbamate pesticide carbofuran causes neurophysiological and neurobehavioral deficits in rodents and humans and therefore poses serious health hazards around the world. Previously, we reported that gestational carbofuran exposure has detrimental effects on hippocampal neurogenesis, the generation of new neurons from neural stem cells (NSC), in offspring. However, the underlying cellular and molecular mechanisms for carbofuran-impaired neurogenesis remain unknown. Herein, we observed that chronic carbofuran exposure from gestational day 7 to postnatal day 21 altered expression of genes and transcription factors and levels of proteins involved in neurogenesis and the TGF-ß pathway (i.e. TGF-ß; SMAD-2, -3, and -7; and SMURF-2) in the rat hippocampus. We found that carbofuran increases TGF-ß signaling (i.e. increased phosphorylated SMAD-2/3 and reduced SMAD-7 expression) in the hippocampus, which reduced NSC proliferation because of increased p21 levels and reduced cyclin D1 levels. Moreover, the carbofuran-altered TGF-ß signaling impaired neuronal differentiation (BrdU/DCX+ and BrdU/NeuN+ cells) and increased apoptosis and neurodegeneration in the hippocampus. Blockade of the TGF-ß pathway with the specific inhibitor SB431542 and via SMAD-3 siRNA prevented carbofuran-mediated inhibition of neurogenesis in both hippocampal NSC cultures and the hippocampus, suggesting the specific involvement of this pathway. Of note, both in vitro and in vivo studies indicated that TGF-ß pathway attenuation reverses carbofuran's inhibitory effects on neurogenesis and associated learning and memory deficits. These results suggest that carbofuran inhibits NSC proliferation and neuronal differentiation by altering TGF-ß signaling. Therefore, we conclude that TGF-ß may represent a potential therapeutic target against carbofuran-mediated neurotoxicity and neurogenesis disruption.


Subject(s)
Carbofuran/pharmacology , Hippocampus/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Smad3 Protein/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Doublecortin Protein , Female , Hippocampus/cytology , Hippocampus/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Signal Transduction
5.
Int J Mol Sci ; 19(10)2018 Oct 14.
Article in English | MEDLINE | ID: mdl-30322209

ABSTRACT

BACKGROUND: Throughout history, menopause has been regarded as a transition in a woman's life. With the increase in life expectancy, women now spend more than a third of their lives in menopause. During these years, women may experience intolerable symptoms both physically and mentally, leading them to seek clinical advice. It is imperative for healthcare providers to improve the quality of life by reducing bothersome menopausal symptoms and preventing disorders such as osteoporosis and atherosclerosis. The current treatment in the form of hormone replacement therapy (HRT) is sometimes inadequate with several limitations and adverse effects. Objective and rationale: The current review aims to discuss the need, efficacy, and limitations of current HRT; the role of other ovarian hormones, and where we stand in comparison with ovary-in situ; and finally, explore towards the preparation of an HRT model by regeneration of ovaries tissues through stem cells which can replicate a functional ovary. SEARCH METHODS: Four electronic databases (MEDLINE, Embase, Web of Science and CINAHL) were searched from database inception until 26 April 2018, using a combination of relevant controlled vocabulary terms and free-text terms related to 'menopause', 'hormone replacement therapy', 'ovary regeneration', 'stem cells' and 'ovarian transplantation'. OUTCOMES: We present a synthesis of the existing data on the efficacy and limitations of HRT. HRT is far from adequate in postmenopausal women with symptoms of hormone deprivation as it fails to deliver all hormones secreted by naïve ovarian tissue. Moreover, the pharmacokinetics of synthetic hormones makes them substantially different from natural ones. Not only does the number and type of hormones given in HRT matter, but the route of delivering and their release in circulation are also imperative. The hormones are delivered either orally or topically in a non-physiological uniform manner, which brings along with it several side effects. These identify the need for a hormone delivery system which replicates, integrates and reacts as per the requirement of the female body. Wider implications: The review outlines the strengths and weaknesses of HRT and highlights the potential areas for future research. There is a tremendous potential for research in this field to understand the collective roles of the various ovarian hormones and to devise an auto-regulated hormone delivery system which replicates the normal physiology. Its clinical applications can prove to be transformative for postmenopausal women helping them to lead a healthy and productive life.


Subject(s)
Hormone Replacement Therapy/methods , Ovary/cytology , Stem Cells/cytology , Female , Humans , Postmenopause , Quality of Life , Tissue Engineering
6.
J Biol Chem ; 291(31): 15923-39, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27252377

ABSTRACT

The regulatory dynamics of mitochondria comprises well orchestrated distribution and mitochondrial turnover to maintain the mitochondrial circuitry and homeostasis inside the cells. Several pieces of evidence suggested impaired mitochondrial dynamics and its association with the pathogenesis of neurodegenerative disorders. We found that chronic exposure of synthetic xenoestrogen bisphenol A (BPA), a component of consumer plastic products, impaired autophagy-mediated mitochondrial turnover, leading to increased oxidative stress, mitochondrial fragmentation, and apoptosis in hippocampal neural stem cells (NSCs). It also inhibited hippocampal derived NSC proliferation and differentiation, as evident by the decreased number of BrdU- and ß-III tubulin-positive cells. All these effects were reversed by the inhibition of oxidative stress using N-acetyl cysteine. BPA up-regulated the levels of Drp-1 (dynamin-related protein 1) and enhanced its mitochondrial translocation, with no effect on Fis-1, Mfn-1, Mfn-2, and Opa-1 in vitro and in the hippocampus. Moreover, transmission electron microscopy studies suggested increased mitochondrial fission and accumulation of fragmented mitochondria and decreased elongated mitochondria in the hippocampus of the rat brain. Impaired mitochondrial dynamics by BPA resulted in increased reactive oxygen species and malondialdehyde levels, disruption of mitochondrial membrane potential, and ATP decline. Pharmacological (Mdivi-1) and genetic (Drp-1siRNA) inhibition of Drp-1 reversed BPA-induced mitochondrial dysfunctions, fragmentation, and apoptosis. Interestingly, BPA-mediated inhibitory effects on NSC proliferation and neuronal differentiations were also mitigated by Drp-1 inhibition. On the other hand, Drp-1 inhibition blocked BPA-mediated Drp-1 translocation, leading to decreased apoptosis of NSC. Overall, our studies implicate Drp-1 as a potential therapeutic target against BPA-mediated impaired mitochondrial dynamics and neurodegeneration in the hippocampus.


Subject(s)
Benzhydryl Compounds/toxicity , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dynamins/metabolism , Hippocampus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Neural Stem Cells/metabolism , Phenols/toxicity , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Hippocampus/pathology , Male , Mitochondria/pathology , Neural Stem Cells/pathology , Protein Transport/drug effects , Rats , Rats, Wistar
7.
Biochem Biophys Res Commun ; 483(4): 1166-1177, 2017 02 19.
Article in English | MEDLINE | ID: mdl-27514452

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and they serve to be a promising therapeutic target for several neurodegenerative disorders, which includes Parkinson disease, Alzheimer's disease, Huntington disease and Amyotrophic Lateral Sclerosis. PPARs play an important role in the downregulation of mitochondrial dysfunction, proteasomal dysfunction, oxidative stress, and neuroinflammation, which are the major causes of the pathogenesis of neurodegenerative disorders. In this review, we discuss about the role of PPARs as therapeutic targets in neurodegenerative disorders. Several experimental approaches suggest potential application of PPAR agonist as well as antagonist in the treatment of neurodegenerative disorders. Several epidemiological studies found that the regular usage of PPAR activating non-steroidal anti-inflammatory drugs is effective in decreasing the progression of neurodegenerative diseases including PD and AD. We also reviewed the neuroprotective effects of PPAR agonists and associated mechanism of action in several neurodegenerative disorders both in vitro as well as in vivo animal models.


Subject(s)
Neurodegenerative Diseases/therapy , Peroxisome Proliferator-Activated Receptors/drug effects , Animals , Calcium/metabolism , Homeostasis , Humans , Inflammation/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Peroxisome Proliferator-Activated Receptors/metabolism
8.
Crit Care Med ; 45(9): e877-e915, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28816851

ABSTRACT

OBJECTIVE: To provide ICU clinicians with evidence-based guidance on safe medication use practices for the critically ill. DATA SOURCES: PubMed, Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, CINAHL, Scopus, and ISI Web of Science for relevant material to December 2015. STUDY SELECTION: Based on three key components: 1) environment and patients, 2) the medication use process, and 3) the patient safety surveillance system. The committee collectively developed Population, Intervention, Comparator, Outcome questions and quality of evidence statements pertaining to medication errors and adverse drug events addressing the key components. A total of 34 Population, Intervention, Comparator, Outcome questions, five quality of evidence statements, and one commentary on disclosure was developed. DATA EXTRACTION: Subcommittee members were assigned selected Population, Intervention, Comparator, Outcome questions or quality of evidence statements. Subcommittee members completed their Grading of Recommendations Assessment, Development, and Evaluation of the question with his/her quality of evidence assessment and proposed strength of recommendation, then the draft was reviewed by the relevant subcommittee. The subcommittee collectively reviewed the evidence profiles for each question they developed. After the draft was discussed and approved by the entire committee, then the document was circulated among all members for voting on the quality of evidence and strength of recommendation. DATA SYNTHESIS: The committee followed the principles of the Grading of Recommendations Assessment, Development, and Evaluation system to determine quality of evidence and strength of recommendations. CONCLUSIONS: This guideline evaluates the ICU environment as a risk for medication-related events and the environmental changes that are possible to improve safe medication use. Prevention strategies for medication-related events are reviewed by medication use process node (prescribing, distribution, administration, monitoring). Detailed considerations to an active surveillance system that includes reporting, identification, and evaluation are discussed. Also, highlighted is the need for future research for safe medication practices that is specific to critically ill patients.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/prevention & control , Intensive Care Units/organization & administration , Medication Errors/prevention & control , Medication Systems, Hospital/organization & administration , Body Weights and Measures , Checklist/standards , Clinical Protocols/standards , Decision Support Systems, Clinical/organization & administration , Disclosure , Documentation/standards , Dose-Response Relationship, Drug , Drug Labeling/methods , Electronic Data Processing , Environment , Evidence-Based Practice , Humans , Infusion Pumps , Inservice Training , Intensive Care Units/standards , Intensive Care Units, Pediatric/organization & administration , Medical Order Entry Systems/organization & administration , Medication Reconciliation/organization & administration , Medication Systems, Hospital/standards , Organizational Culture , Patient Care Bundles/standards , Patient Handoff/standards , Patient Participation , Risk Factors , Software Design
10.
J Biol Chem ; 290(47): 28540-28558, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26420483

ABSTRACT

Neurogenesis involves generation of new neurons through finely tuned multistep processes, such as neural stem cell (NSC) proliferation, migration, differentiation, and integration into existing neuronal circuitry in the dentate gyrus of the hippocampus and subventricular zone. Adult hippocampal neurogenesis is involved in cognitive functions and altered in various neurodegenerative disorders, including Alzheimer disease (AD). Ethosuximide (ETH), an anticonvulsant drug is used for the treatment of epileptic seizures. However, the effects of ETH on adult hippocampal neurogenesis and the underlying cellular and molecular mechanism(s) are yet unexplored. Herein, we studied the effects of ETH on rat multipotent NSC proliferation and neuronal differentiation and adult hippocampal neurogenesis in an amyloid ß (Aß) toxin-induced rat model of AD-like phenotypes. ETH potently induced NSC proliferation and neuronal differentiation in the hippocampus-derived NSC in vitro. ETH enhanced NSC proliferation and neuronal differentiation and reduced Aß toxin-mediated toxicity and neurodegeneration, leading to behavioral recovery in the rat AD model. ETH inhibited Aß-mediated suppression of neurogenic and Akt/Wnt/ß-catenin pathway gene expression in the hippocampus. ETH activated the PI3K·Akt and Wnt·ß-catenin transduction pathways that are known to be involved in the regulation of neurogenesis. Inhibition of the PI3K·Akt and Wnt·ß-catenin pathways effectively blocked the mitogenic and neurogenic effects of ETH. In silico molecular target prediction docking studies suggest that ETH interacts with Akt, Dkk-1, and GSK-3ß. Our findings suggest that ETH stimulates NSC proliferation and differentiation in vitro and adult hippocampal neurogenesis via the PI3K·Akt and Wnt·ß-catenin signaling.


Subject(s)
Alzheimer Disease/chemically induced , Amyloid beta-Peptides/toxicity , Ethosuximide/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cognition Disorders/chemically induced , Cognition Disorders/enzymology , Cognition Disorders/metabolism , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/metabolism , Hippocampus/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Wnt Proteins/metabolism , beta Catenin/metabolism
12.
J Biol Chem ; 290(34): 21163-21184, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26139607

ABSTRACT

The human health hazards related to persisting use of bisphenol-A (BPA) are well documented. BPA-induced neurotoxicity occurs with the generation of oxidative stress, neurodegeneration, and cognitive dysfunctions. However, the cellular and molecular mechanism(s) of the effects of BPA on autophagy and association with oxidative stress and apoptosis are still elusive. We observed that BPA exposure during the early postnatal period enhanced the expression and the levels of autophagy genes/proteins. BPA treatment in the presence of bafilomycin A1 increased the levels of LC3-II and SQSTM1 and also potentiated GFP-LC3 puncta index in GFP-LC3-transfected hippocampal neural stem cell-derived neurons. BPA-induced generation of reactive oxygen species and apoptosis were mitigated by a pharmacological activator of autophagy (rapamycin). Pharmacological (wortmannin and bafilomycin A1) and genetic (beclin siRNA) inhibition of autophagy aggravated BPA neurotoxicity. Activation of autophagy against BPA resulted in intracellular energy sensor AMP kinase (AMPK) activation, increased phosphorylation of raptor and acetyl-CoA carboxylase, and decreased phosphorylation of ULK1 (Ser-757), and silencing of AMPK exacerbated BPA neurotoxicity. Conversely, BPA exposure down-regulated the mammalian target of rapamycin (mTOR) pathway by phosphorylation of raptor as a transient cell's compensatory mechanism to preserve cellular energy pool. Moreover, silencing of mTOR enhanced autophagy, which further alleviated BPA-induced reactive oxygen species generation and apoptosis. BPA-mediated neurotoxicity also resulted in mitochondrial loss, bioenergetic deficits, and increased PARKIN mitochondrial translocation, suggesting enhanced mitophagy. These results suggest implication of autophagy against BPA-mediated neurodegeneration through involvement of AMPK and mTOR pathways. Hence, autophagy, which arbitrates cell survival and demise during stress conditions, requires further assessment to be established as a biomarker of xenoestrogen exposure.


Subject(s)
Autophagy/drug effects , Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Hippocampus/drug effects , Neurons/drug effects , Phenols/toxicity , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Androstadienes/pharmacology , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Beclin-1 , Benzhydryl Compounds/antagonists & inhibitors , Environmental Pollutants/antagonists & inhibitors , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Macrolides/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Phenols/antagonists & inhibitors , Primary Cell Culture , Protein Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Wortmannin
13.
Glia ; 64(7): 1252-64, 2016 07.
Article in English | MEDLINE | ID: mdl-27189737

ABSTRACT

The astrocytic GLT-1 (or EAAT2) is the major glutamate transporter for clearing synaptic glutamate. While the diffusion dynamics of neurotransmitter receptors at the neuronal surface are well understood, far less is known regarding the surface trafficking of transporters in subcellular domains of the astrocyte membrane. Here, we have used live-cell imaging to study the mechanisms regulating GLT-1 surface diffusion in astrocytes in dissociated and brain slice cultures. Using GFP-time lapse imaging, we show that GLT-1 forms stable clusters that are dispersed rapidly and reversibly upon glutamate treatment in a transporter activity-dependent manner. Fluorescence recovery after photobleaching and single particle tracking using quantum dots revealed that clustered GLT-1 is more stable than diffuse GLT-1 and that glutamate increases GLT-1 surface diffusion in the astrocyte membrane. Interestingly, the two main GLT-1 isoforms expressed in the brain, GLT-1a and GLT-1b, are both found to be stabilized opposed to synapses under basal conditions, with GLT-1b more so. GLT-1 surface mobility is increased in proximity to activated synapses and alterations of neuronal activity can bidirectionally modulate the dynamics of both GLT-1 isoforms. Altogether, these data reveal that astrocytic GLT-1 surface mobility, via its transport activity, is modulated during neuronal firing, which may be a key process for shaping glutamate clearance and glutamatergic synaptic transmission. GLIA 2016;64:1252-1264.


Subject(s)
Astrocytes/physiology , Biological Transport/physiology , Cerebral Cortex/cytology , Excitatory Amino Acid Transporter 2/metabolism , Neurons/physiology , 4-Aminopyridine/pharmacology , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Astrocytes/drug effects , Biological Transport/genetics , Cells, Cultured , Coculture Techniques , Embryo, Mammalian , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/pharmacology , Hippocampus/cytology , Neurons/drug effects , Organ Culture Techniques , Potassium Channel Blockers/pharmacology , Rats , Rats, Transgenic , Tetrodotoxin/pharmacology
14.
Indian J Chest Dis Allied Sci ; 57(4): 235-8, 2015.
Article in English | MEDLINE | ID: mdl-27164732

ABSTRACT

We report a case of tumourlets of the lung associated with carcinoid and neuroendocrine cell hyperplasia, found incidentally in a 30-year-old woman, who underwent bullectomy for pneumothorax. These lesions are histologically similar to carcinoid, but differ in molecular pathogenesis about which little is known. Their nature and significance is debated. Here, we point out the importance of histological, clinical, and diagnostic aspects and follow-up to have evidence of eventual malignant evolution.


Subject(s)
Carcinoid Tumor , Lung Neoplasms , Lung/pathology , Pneumothorax , Adult , Carcinoid Tumor/complications , Carcinoid Tumor/pathology , Carcinoid Tumor/therapy , Disease Management , Female , Humans , Lung Neoplasms/complications , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Pneumonectomy/methods , Pneumothorax/diagnosis , Pneumothorax/etiology , Pneumothorax/surgery
15.
Learn Mem ; 21(7): 351-62, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24939839

ABSTRACT

It is well established that fear memory formation requires de novo gene transcription in the amygdala. We provide evidence that epigenetic mechanisms in the form of histone lysine methylation in the lateral amygdala (LA) are regulated by NMDA receptor (NMDAR) signaling and involved in gene transcription changes necessary for fear memory consolidation. Here we found increases in histone H3 lysine 9 dimethylation (H3K9me2) levels in the LA at 1 h following auditory fear conditioning, which continued to be temporally regulated up to 25 h following behavioral training. Additionally, we demonstrate that inhibiting the H3K9me2 histone lysine methyltransferase G9a (H/KMTs-G9a) in the LA impaired fear memory, while blocking the H3K9me2 histone lysine demethylase LSD1 (H/KDM-LSD1) enhanced fear memory, suggesting that H3K9me2 in the LA can bidirectionally regulate fear memory formation. Furthermore, we show that NMDAR activity differentially regulated the recruitment of H/KMT-G9a, H/KDM-LSD1, and subsequent H3K9me2 levels at a target gene promoter. This was largely regulated by GluN2B- but not GluN2A-containing NMDARs via ERK activation. Moreover, fear memory deficits associated with NMDAR or ERK blockade were successfully rescued through pharmacologically inhibiting LSD1, suggesting that enhancements of H3K9me2 levels within the LA can rescue fear memory impairments that result from hypofunctioning NMDARs or loss of ERK signaling. Together, the present study suggests that histone lysine methylation regulation in the LA via NMDAR-ERK-dependent signaling is involved in fear memory formation.


Subject(s)
Basolateral Nuclear Complex/physiology , Fear/physiology , Histones/metabolism , Memory, Long-Term/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Association Learning/drug effects , Association Learning/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , MAP Kinase Signaling System/physiology , Male , Memory Disorders/genetics , Memory Disorders/physiopathology , Memory, Long-Term/drug effects , Methylation , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Reflex, Startle/physiology
16.
Hum Mol Genet ; 21(15): 3474-88, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22589249

ABSTRACT

Huntington's disease (HD) is an incurable neurological disorder caused by an abnormal glutamine repeat expansion in the huntingtin (Htt) protein. In the present studies, we investigated the role of Transducers of Regulated cAMP response element-binding (CREB) protein activity (TORCs) in HD, since TORCs play an important role in the expression of the transcriptional co-regulator peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), whose expression is impaired in HD. We found significantly decreased TORC1 expression levels in STHdhQ111 cells expressing mutant Htt, in the striatum of NLS-N171-82Q, R6/2 and HdhQ111 HD transgenic mice and in postmortem striatal tissue from HD patients. TORC1 overexpression in wild-type (WT) and Htt striatal cells increased CREB mRNA and protein levels, PGC-1α promoter activity, mRNA expression of the PGC-1α, NRF-1, Tfam and CytC genes, mitochondrial DNA content, mitochondrial activity and mitochondrial membrane potential. TORC1 overexpression also increased the resistance of striatal cells to 3-nitropropionic (3-NP) acid-mediated toxicity. In cultured WT and mutant Htt striatal cells, small hairpin RNA-mediated TORC1 knockdown resulted in decreased PGC-1α expression and increased susceptibility to 3-NP-induced toxicity. Overexpression of PGC-1α partially prevented TORC1 knockdown-mediated increased susceptibility of Htt striatal cells to 3-NP. Specific knockdown of TORC1 in the striatum of NLS-N171-82Q HD transgenic mice induced neurodegeneration. Lastly, knockdown of Htt prevents transcriptional repression of TORC1 and CREB in Htt striatal cells. These findings show that impaired expression and function of TORC1, which results in a reduction in PGC-1α, plays an important role in mitochondrial dysfunction in HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic
17.
Biochem Soc Trans ; 42(5): 1302-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25233407

ABSTRACT

Astrocytes exhibit cellular excitability through variations in their intracellular calcium (Ca²âº) levels in response to synaptic activity. Astrocyte Ca²âº elevations can trigger the release of neuroactive substances that can modulate synaptic transmission and plasticity, hence promoting bidirectional communication with neurons. Intracellular Ca²âº dynamics can be regulated by several proteins located in the plasma membrane, within the cytosol and by intracellular organelles such as mitochondria. Spatial dynamics and strategic positioning of mitochondria are important for matching local energy provision and Ca²âº buffering requirements to the demands of neuronal signalling. Although relatively unresolved in astrocytes, further understanding the role of mitochondria in astrocytes may reveal more about the complex bidirectional relationship between astrocytes and neurons in health and disease. In the present review, we discuss some recent insights regarding mitochondrial function, transport and turnover in astrocytes and highlight some important questions that remain to be answered.


Subject(s)
Astrocytes/metabolism , Mitochondrial Dynamics , Models, Biological , Animals , Astrocytes/cytology , Astrocytes/pathology , Calcium Signaling , Humans , Mitochondria/metabolism , Mitochondria/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
18.
Pediatr Crit Care Med ; 15(2): e38-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24201855

ABSTRACT

OBJECTIVE: The revised guidelines for the determination of brain death in infants and children stress that apnea testing is an integral component in determining brain death based on clinical criteria. Unfortunately, these guidelines provide no process for apnea testing during the determination of brain death in patients supported on venoarterial extracorporeal membrane oxygenation. We review three pediatric patients supported on venoarterial extracorporeal membrane oxygenation who underwent apnea testing during their brain death evaluation. This is the only published report to elucidate a reliable, successful method for apnea testing in pediatric patients supported on venoarterial extracorporeal membrane oxygenation. DESIGN: Retrospective case series. SETTING: Two tertiary care PICUs in university teaching hospitals. PATIENTS: Three pediatric patients supported by venoarterial extracorporeal membrane oxygenation after cardiopulmonary arrest. INTERVENTIONS: After neurologic examinations demonstrated cessation of brain function in accordance with current pediatric brain death guidelines, apnea testing was performed on each child while supported on venoarterial extracorporeal membrane oxygenation. MEASUREMENTS AND MAIN RESULTS: In two of the three cases, the patients remained hemodynamically stable with normal oxygen saturations as venoarterial extracorporeal membrane oxygenation sweep gas was weaned and apnea testing was undertaken. Apnea testing demonstrating no respiratory effort was successfully completed in these two cases. The third patient became hemodynamically unstable, invalidating the apnea test. CONCLUSIONS: Apnea testing on venoarterial extracorporeal membrane oxygenation can be successfully undertaken in the evaluation of brain death. We provide a suggested protocol for apnea testing while on venoarterial extracorporeal membrane oxygenation that is consistent with the updated pediatric brain death guidelines. This is the only published report to elucidate a reliable, successful method for apnea testing in pediatric patients supported on venoarterial extracorporeal membrane oxygenation.


Subject(s)
Apnea/diagnosis , Brain Death/diagnosis , Extracorporeal Membrane Oxygenation/methods , Adolescent , Child , Child, Preschool , Hospitals, University , Humans , Infant , Intensive Care Units, Pediatric , Pediatrics , Retrospective Studies
19.
J Neurosci ; 32(16): 5440-53, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22514307

ABSTRACT

Learning triggers alterations in gene transcription in brain regions such as the hippocampus and the entorhinal cortex (EC) that are necessary for long-term memory (LTM) formation. Here, we identify an essential role for the G9a/G9a-like protein (GLP) lysine dimethyltransferase complex and the histone H3 lysine 9 dimethylation (H3K9me2) marks it catalyzes, in the transcriptional regulation of genes in area CA1 of the rat hippocampus and the EC during memory consolidation. Contextual fear learning increased global levels of H3K9me2 in area CA1 and the EC, with observable changes at the Zif268, DNMT3a, BDNF exon IV, and cFOS gene promoters, which occurred in concert with mRNA expression. Inhibition of G9a/GLP in the EC, but not in the hippocampus, enhanced contextual fear conditioning relative to control animals. The inhibition of G9a/GLP in the EC induced several histone modifications that include not only methylation but also acetylation. Surprisingly, we found that downregulation of G9a/GLP activity in the EC enhanced H3K9me2 in area CA1, resulting in transcriptional silencing of the non-memory permissive gene COMT in the hippocampus. In addition, synaptic plasticity studies at two distinct EC-CA1 cellular pathways revealed that G9a/GLP activity is critical for hippocampus-dependent long-term potentiation initiated in the EC via the perforant pathway, but not the temporoammonic pathway. Together, these data demonstrate that G9a/GLP differentially regulates gene transcription in the hippocampus and the EC during memory consolidation. Furthermore, these findings support the possibility of a role for G9a/GLP in the regulation of cellular and molecular cross talk between these two brain regions during LTM formation.


Subject(s)
Entorhinal Cortex/enzymology , Gene Silencing/physiology , Hippocampus/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Memory/physiology , Transcriptional Activation/physiology , Analysis of Variance , Animals , Azepines/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Chromatin Immunoprecipitation , Conditioning, Psychological/physiology , Cues , DNA (Cytosine-5-)-Methyltransferases , DNA Methyltransferase 3A , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Fear , Gene Silencing/drug effects , Hippocampus/cytology , Hippocampus/physiology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histones/metabolism , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Memory/drug effects , Methylation , Patch-Clamp Techniques , Polymers , Proto-Oncogene Proteins c-fyn/metabolism , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Transcriptional Activation/drug effects
20.
Plants (Basel) ; 12(4)2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36840279

ABSTRACT

Syzygium cumini L. (ver Jamun; BlackBerry) is a native, evergreen multipurpose tree species of India. Besides being a fruit tree and for agroforestry in different regions, it is medicinally important too. This study aimed to determine genetic diversity using molecular and phytochemical markers in sixteen genotypes of Indian S. cumini from different agro-ecological zones. The present study used a combination of ISSR markers and the HPLC technique to explore these genotypes. The results showed a wide genetic diversity range based on the similarity coefficient values observed in S. cumini sixteen accessions from different sites. Four primary phenolic acids were discovered in all the accessions; caffeic acid (CA) was found in high concentrations. The intraspecific association between molecular and phytochemical characteristics was the primary goal of this investigation. By employing gene-specific markers for the route of secondary metabolites (polyphenols) production, it further investigated the progressive research of diversity analysis of polyphenol content in S. cumini accessions, which may also expand its nutraceutical and pharmaceutical utilization.

SELECTION OF CITATIONS
SEARCH DETAIL