Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Lancet Digit Health ; 6(2): e93-e104, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38278619

ABSTRACT

BACKGROUND: Multicentre training could reduce biases in medical artificial intelligence (AI); however, ethical, legal, and technical considerations can constrain the ability of hospitals to share data. Federated learning enables institutions to participate in algorithm development while retaining custody of their data but uptake in hospitals has been limited, possibly as deployment requires specialist software and technical expertise at each site. We previously developed an artificial intelligence-driven screening test for COVID-19 in emergency departments, known as CURIAL-Lab, which uses vital signs and blood tests that are routinely available within 1 h of a patient's arrival. Here we aimed to federate our COVID-19 screening test by developing an easy-to-use embedded system-which we introduce as full-stack federated learning-to train and evaluate machine learning models across four UK hospital groups without centralising patient data. METHODS: We supplied a Raspberry Pi 4 Model B preloaded with our federated learning software pipeline to four National Health Service (NHS) hospital groups in the UK: Oxford University Hospitals NHS Foundation Trust (OUH; through the locally linked research University, University of Oxford), University Hospitals Birmingham NHS Foundation Trust (UHB), Bedfordshire Hospitals NHS Foundation Trust (BH), and Portsmouth Hospitals University NHS Trust (PUH). OUH, PUH, and UHB participated in federated training, training a deep neural network and logistic regressor over 150 rounds to form and calibrate a global model to predict COVID-19 status, using clinical data from patients admitted before the pandemic (COVID-19-negative) and testing positive for COVID-19 during the first wave of the pandemic. We conducted a federated evaluation of the global model for admissions during the second wave of the pandemic at OUH, PUH, and externally at BH. For OUH and PUH, we additionally performed local fine-tuning of the global model using the sites' individual training data, forming a site-tuned model, and evaluated the resultant model for admissions during the second wave of the pandemic. This study included data collected between Dec 1, 2018, and March 1, 2021; the exact date ranges used varied by site. The primary outcome was overall model performance, measured as the area under the receiver operating characteristic curve (AUROC). Removable micro secure digital (microSD) storage was destroyed on study completion. FINDINGS: Clinical data from 130 941 patients (1772 COVID-19-positive), routinely collected across three hospital groups (OUH, PUH, and UHB), were included in federated training. The evaluation step included data from 32 986 patients (3549 COVID-19-positive) attending OUH, PUH, or BH during the second wave of the pandemic. Federated training of a global deep neural network classifier improved upon performance of models trained locally in terms of AUROC by a mean of 27·6% (SD 2·2): AUROC increased from 0·574 (95% CI 0·560-0·589) at OUH and 0·622 (0·608-0·637) at PUH using the locally trained models to 0·872 (0·862-0·882) at OUH and 0·876 (0·865-0·886) at PUH using the federated global model. Performance improvement was smaller for a logistic regression model, with a mean increase in AUROC of 13·9% (0·5%). During federated external evaluation at BH, AUROC for the global deep neural network model was 0·917 (0·893-0·942), with 89·7% sensitivity (83·6-93·6) and 76·6% specificity (73·9-79·1). Site-specific tuning of the global model did not significantly improve performance (change in AUROC <0·01). INTERPRETATION: We developed an embedded system for federated learning, using microcomputing to optimise for ease of deployment. We deployed full-stack federated learning across four UK hospital groups to develop a COVID-19 screening test without centralising patient data. Federation improved model performance, and the resultant global models were generalisable. Full-stack federated learning could enable hospitals to contribute to AI development at low cost and without specialist technical expertise at each site. FUNDING: The Wellcome Trust, University of Oxford Medical and Life Sciences Translational Fund.


Subject(s)
COVID-19 , Secondary Care , Humans , Artificial Intelligence , Privacy , State Medicine , COVID-19/diagnosis , Hospitals , United Kingdom
2.
Neurochem Int ; 146: 105022, 2021 06.
Article in English | MEDLINE | ID: mdl-33746005

ABSTRACT

Inflammation plays a role in neuropathology. We hypothesised that inflammation, induced by a single intraperitoneal injection of lipopolysaccharide (LPS), would induce long-term changes in the regulation of tyrosine hydroxylase (TH) in the rat midbrain. The level of 12 cytokines was initially analysed from one day to six months after LPS injection to confirm that peripheral inflammation led to neuroinflammatory changes in the midbrain. In the substantia nigra (SN), the levels of 8 of the 12 measured cytokines was significantly increased at one day. Granulocyte-macrophage colony-stimulating factor showed a threefold increased level at 6 months. The ventral tegmental area (VTA) showed a completely different pattern, with no increases in the levels of the 12 cytokines at one day and no changes beyond one week. TH activity was determined using a tritiated water release assay, TH protein and phosphorylation levels (Ser19, Ser31 and Ser40) were determined using western blotting. TH-specific activity in the SN was unchanged at one day but was substantially increased at one week and one month with no concomitant increase in TH phosphorylation. Substantial changes in TH activation without changes in TH phosphorylation have not previously been observed in the brain in response to a range of stressors. TH-specific activity was increased in the SN, and in the caudate putamen, at 6 months and was associated with increased TH phosphorylation at Ser19 and Ser40 at both locations. TH-specific activity in the VTA showed only a transient increase at day one associated with increased phosphorylation at Ser19 and Ser31 but thereafter showed no changes. This study shows that inflammation induced by LPS generated two distinct long-term changes in TH activity in the SN that are caused by different mechanisms, but there were no long-term changes in the adjacent VTA.


Subject(s)
Inflammation Mediators/metabolism , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/metabolism , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Lipopolysaccharides/toxicity , Male , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Time Factors
3.
Neurobiol Dis ; 130: 104524, 2019 10.
Article in English | MEDLINE | ID: mdl-31276794

ABSTRACT

Tyrosine hydroxylase is the key enzyme controlling the synthesis of the catecholamines including dopamine. The breakdown of dopamine into toxic compounds has been suggested to have a key role in the degeneration of the dopaminergic neurons in Parkinson's disease. Humans are unique in containing four isoforms of tyrosine hydroxylase, but understanding of the role of these isoforms under normal conditions and in disease states is limited. The aim of this work was to determine the level and distribution of the four human isoforms in tissues from healthy controls and patients with Parkinson's disease. The results show that isoform 1 and isoform 2 are the major tyrosine hydroxylase isoforms in human brain, but that tyrosine hydroxylase isoform 2 is more abundant in the substantia nigra than the tyrosine hydroxylase isoform 1. The two minor isoforms, isoform 3 and isoform 4, are expressed at a proportionally higher level in the terminal field regions (caudate and putamen) compared to the substantia nigra. There was a selective loss of tyrosine hydroxylase isoform 1 in Parkinson's disease compared to age-matched controls and a corresponding increase in the proportion of tyrosine hydroxylase isoform 2. Phosphorylation of serine 40 was significantly increased in caudate, putamen and ventral tegmental area, but not in the substantia nigra, in Parkinson's disease brain. These results show a selective sparing of tyrosine hydroxylase isoform 2 in Parkinson's disease. Isoform 2 exhibits a reduced capacity for activation compared to isoform 1, which may account for the selective sparing of cells expressing isoform 2 in Parkinson's disease. Surviving neurons in Parkinson's disease brain exhibit a substantial increase in tyrosine hydroxylase phosphorylation consistent with a compensatory mechanism of increased dopamine synthesis in the terminal field regions.


Subject(s)
Corpus Striatum/metabolism , Parkinson Disease/metabolism , Protein Isoforms/metabolism , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , Dopaminergic Neurons/metabolism , Humans , Phosphorylation
4.
J Cell Biochem ; 120(12): 19730-19737, 2019 12.
Article in English | MEDLINE | ID: mdl-31297896

ABSTRACT

Tyrosine hydroxylase (TH) is the key enzyme that controls the rate of synthesis of the catecholamines. SH-SY5Y cells with stable transfections of either human tyrosine hydroxylase isoform 1 (hTH1) or human tyrosine hydroxylase isoform 4 (hTH4) were used to determined the subcellular distribution of TH protein and phosphorylated TH, under basal conditions and after muscarine stimulation. Muscarine was previously shown to increase the phosphorylation of only serine 19 and serine 40 in hTH1 cells. Under basal conditions, the hTH1 and hTH4 proteins, their serine 19 phosphorylated forms and hTH1 phosphorylated at serine 40 were all similarly distributed; with ~80% in the cytosolic fraction, ~20% in the membrane fraction, and less than 1%, or not detectable, in the nuclear fraction. However, hTH4 phosphorylated at serine 71 had a significantly different distribution with ~65% cytosolic and ~35% membrane associated. Muscarine stimulation led to hTH1 being redistributed from the cytosol and nuclear fractions to the membrane fraction and hTH4 being redistributed from the cytosol to the nuclear fraction. These muscarine stimulated redistributions were not due to TH phosphorylation at serine 19, serine 40, or serine 71 and were most likely due to TH binding to proteins whose phosphorylation was increased by muscarine. This is the first study to show a difference in subcellular distribution between two human TH isoforms under basal and stimulated conditions.


Subject(s)
Tyrosine 3-Monooxygenase/metabolism , Cell Line , Cell Membrane/enzymology , Cytosol/metabolism , Humans , Isoenzymes/metabolism , Muscarine/pharmacology , Phosphorylation , Serine/metabolism , Subcellular Fractions/enzymology , Tyrosine 3-Monooxygenase/genetics
5.
J Neurochem ; 149(6): 706-728, 2019 06.
Article in English | MEDLINE | ID: mdl-30714137

ABSTRACT

Tyrosine hydroxylase (TH) is the rate-limiting enzyme in the synthesis of the catecholamines dopamine, noradrenaline and adrenaline. One of the major mechanisms for controlling the activity of TH is protein phosphorylation. TH is phosphorylated at serine residues 8, 19, 31 and 40. There have been a number of previous reviews focused on TH phosphorylation in vitro and in situ. This review on TH phosphorylation in vivo has three main sections focusing on: (1) the methods used to investigate TH phosphorylation in vivo, including the animals used, the sacrifice procedures, the tissue preparation, the measurement of TH protein levels and TH phosphorylation and the measurement of TH activation. (2) The regulation of TH phosphorylation and its consequences in vivo, including the kinases and phosphatases acting on TH, the stoichiometry of TH phosphorylation, the proteins that bind TH and TH subcellular location. (3) The acute and prolonged TH phosphorylation changes in specific catecholaminergic tissues, including the adrenal medulla, the nigrostriatal pathway and the mesolimbic pathway.


Subject(s)
Tyrosine 3-Monooxygenase/metabolism , Animals , Humans , Phosphorylation
6.
Neuroimmunomodulation ; 25(3): 163-175, 2018.
Article in English | MEDLINE | ID: mdl-30415249

ABSTRACT

OBJECTIVES: Gastrointestinal (GI) inflammation and GI integrity deficits are common comorbidities of neuropsychiatric disorders. Ongoing research suggests that these aberrations may be contributing to heightened immune signals that have the potential to disrupt neuronal homeostasis and exacerbate behavioural deficits. The current study aimed to determine whether the well-characterized animal model of neuropsychopathology, the maternal immune activation (MIA) model, produced GI inflammation and integrity disruptions in association with anxiety-like behaviour. METHODS: Pregnant Wistar rats were exposed to the viral mimetic polyriboinosinic:polyribocytidilic acid (polyI:C) on gestational days (GD) 10 and 19. Evidence of ANS activation, GI inflammation, and GI barrier integrity was assessed in both neonatal (postnatal day, P7) and adult (P84) offspring. Anxiety-like behaviour was assessed at P100. RESULTS: Neonatal MIA offspring exhibited an altered intestinal inflammatory profile and evidence of an increase in lymphoid aggregates. MIA neonates also displayed disruptions to GI barrier tight junction protein mRNA. In addition, adult MIA offspring exhibited an increase in anxiety-like behaviours. CONCLUSION: These results indicate that the MIA rat model, which is well documented to produce behavioural, neurochemical, and neuroanatomical abnormalities, also produces GI inflammation and integrity disruptions. We suggest that this model may be a useful tool to elucidate biological pathways associated with neuropsychiatric disorders.


Subject(s)
Anxiety/chemically induced , Anxiety/immunology , Gastrointestinal Tract/immunology , Inflammation Mediators/immunology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Animals , Anxiety/metabolism , Disease Models, Animal , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Maze Learning/physiology , Poly I-C/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar
7.
Mol Cell Endocrinol ; 470: 295-303, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29183807

ABSTRACT

A poor nutritional environment during early development has long been known to increase disease susceptibility later in life. We have previously shown that rats that are overfed as neonates (i.e. suckled in small litters (4 pups) relative to control conditions (12 pups)) show dysregulated hypothalamic-pituitary-adrenal axis responses to immune stress in adulthood, particularly due to an altered capacity of the adrenal to respond to an immune challenge. Here we hypothesised that neonatal overfeeding similarly affects the sympathomedullary system, testing this by investigating the biochemical function of tyrosine hydroxylase (TH), the first rate-limiting enzyme in the catecholamine synthesis. We also examined changes in adrenal expression of the leptin receptor and in mitogen-activated protein kinase (MAPK) signalling. During the neonatal period, we saw age-dependent changes in TH activity and phosphorylation, with neonatal overfeeding stimulating increased adrenal TH specific activity at postnatal days 7 and 14, along with a compensatory reduction in total TH protein levels. This increased TH activity was maintained into adulthood where neonatally overfed rats exhibited increased adrenal responsiveness 30 min after an immune challenge with lipopolysaccharide, evident in a concomitant increase in TH protein levels and specific activity. Neonatal overfeeding significantly reduced the expression of the leptin receptor in neonatal adrenals at postnatal day 7 and in adult adrenals, but did not affect MAPK signalling. These data suggest neonatal overfeeding alters the capacity of the adrenal to synthesise catecholamines, both acutely and long term, and these effects may be independent of leptin signalling.


Subject(s)
Adrenal Glands/metabolism , Catecholamines/biosynthesis , Feeding Behavior , Animals , Animals, Newborn , Lipopolysaccharides , MAP Kinase Signaling System , Male , Phosphorylation , Rats, Wistar , Receptors, Leptin/metabolism , Tyrosine 3-Monooxygenase/metabolism
8.
Neurochem Int ; 107: 33-42, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28153786

ABSTRACT

Studies in multiple experimental systems show that Ca2+-calmodulin stimulated protein kinase II (CaMKII) is a major mediator of ischaemia-induced cell death and suggest that CaMKII would be a good target for neuroprotective therapies in acute treatment of stroke. However, as CaMKII regulates many cellular processes in many tissues any clinical treatment involving the inhibition of CaMKII would need to be able to specifically target the functions of ischaemia-activated CaMKII. In this review we summarise new developments in our understanding of the molecular mechanisms involved in ischaemia-induced CaMKII-mediated cell death that have identified ways in which such specificity of CaMKII inhibition after stroke could be achieved. We also review the mechanisms and phases of tissue damage in ischaemic stroke to identify where and when CaMKII-mediated mechanisms may be involved.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/prevention & control , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Neuroprotective Agents/administration & dosage , Stroke/enzymology , Stroke/prevention & control , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Death/drug effects , Cell Death/physiology , Drug Delivery Systems/trends , Humans , Neuroprotection/drug effects , Neuroprotection/physiology , Protein Kinase Inhibitors/administration & dosage , Protein Structure, Secondary
9.
Brain Behav Immun ; 62: 124-136, 2017 May.
Article in English | MEDLINE | ID: mdl-28088642

ABSTRACT

Systemic inflammation induces transient or permanent dysfunction in the brain by exposing it to soluble inflammatory mediators. The receptor for advanced glycation endproducts (RAGE) binds to distinct ligands mediating and increasing inflammatory processes. In this study we used an LPS-induced systemic inflammation model in rats to investigate the effect of blocking RAGE in serum, liver, cerebrospinal fluid (CSF) and brain (striatum, prefrontal cortex, ventral tegmental area and substantia nigra). Intraperitoneal injection of RAGE antibody (50µg/kg) was followed after 1h by a single LPS (5mg/kg) intraperitoneal injection. Twenty-four hours later, tissues were isolated for analysis. RAGE antibody reduced LPS-induced inflammatory effects in both serum and liver; the levels of proinflammatory cytokines (TNF-α, IL-1ß) were decreased and the phosphorylation/activation of RAGE downstream targets (ERK1/2, IκB and p65) in liver were significantly attenuated. RAGE antibody prevented LPS-induced effects on TNF-α and IL-1ß in CSF. In striatum, RAGE antibody inhibited increases in IL-1ß, Iba-1, GFAP, phospho-ERK1/2 and phospho-tau (ser202), as well as the decrease in synaptophysin levels. These effects were caused by systemic RAGE inhibition, as RAGE antibody did not cross the blood-brain barrier. RAGE antibody also prevented striatal lipoperoxidation and activation of mitochondrial complex II. In conclusion, blockade of RAGE is able to inhibit inflammatory responses induced by LPS in serum, liver, CSF and brain.


Subject(s)
Antibodies/pharmacology , Corpus Striatum/drug effects , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Liver/drug effects , Receptor for Advanced Glycation End Products/immunology , Animals , Antibodies/therapeutic use , Corpus Striatum/metabolism , Cytokines/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Liver/metabolism , Male , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
10.
Sci Rep ; 7: 40475, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071709

ABSTRACT

Neonatal immune challenge with the bacterial mimetic lipopolysaccharide has the capacity to generate long-term changes in the brain. Neonatal rats were intraperitoneally injected with lipopolysaccharide (0.05 mg/kg) on postnatal day (PND) 3 and again on PND 5. The activation state of tyrosine hydroxylase (TH) was measured in the locus coeruleus, ventral tegmental area and substantia nigra on PND 85. In the locus coeruleus there was an approximately four-fold increase in TH activity. This was accompanied by a significant increase in TH protein together with increased phosphorylation of all three serine residues in the N-terminal region of TH. In the ventral tegmental area, a significant increase in TH activity and increased phosphorylation of the serine 40 residue was seen. Neonatal lipopolysaccharide had no effect on TH activation in the substantia nigra. These results indicate the capacity of a neonatal immune challenge to generate long-term changes in the activation state of TH, in particular in the locus coeruleus. Overall, the current results demonstrate the enduring outcomes of a neonatal immune challenge on specific brain catecholaminergic regions associated with catecholamine synthesis. This highlights a novel mechanism for long-term physiological and behavioural alterations induced by this model.


Subject(s)
Catecholamines/metabolism , Lipopolysaccharides/pharmacology , Neurons/metabolism , Animals , Animals, Newborn , Calcium-Binding Proteins , Glial Fibrillary Acidic Protein/metabolism , Locus Coeruleus/metabolism , Microfilament Proteins , Neurons/drug effects , Phosphorylation/drug effects , Rats, Wistar , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/metabolism
11.
Neurochem Int ; 104: 6-10, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28065796

ABSTRACT

Ischaemia/excitotoxicity produces persistent activation of CaMKII (Ca2+-calmodulin stimulated protein kinase II) that initiates cell death. This study investigated the involvement of CaMKII phosphorylation at T286 and T253 in producing this persistent activation. In T286A-αCaMKII transgenic mice that lack the ability to phosphorylate αCaMKII at T286, transient occlusion of the middle cerebral artery for 90 min resulted in no significant difference in infarct size compared to normal littermate controls. Overexpression of the phospho-mimic mutant T286D-αCaMKII in differentiated neuroblastoma cell lines did not enhance excitotoxicity-induced cell death compared to overexpression of wild type αCaMKII. By contrast, overexpression of the phospho-mimic mutant T253D-αCaMKII significantly enhanced excitotoxicity-induced cell death whereas overexpression of the phospho-null mutant T253V-αCaMKII produced no enhancement. These results indicate that T286 phosphorylation does not play a significant role in ischaemia/excitotoxicity induced CaMKII-mediated cell death and suggest that T253 phosphorylation is required to produce the persistent activation of CaMKII involved in ischaemia/excitotoxicity induced cell death.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Ischemia/metabolism , Animals , Calcium/metabolism , Cell Death , Mice, Inbred C57BL , Neuroblastoma/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation
12.
J Cell Biochem ; 118(8): 2096-2107, 2017 08.
Article in English | MEDLINE | ID: mdl-27958651

ABSTRACT

Immune activation can alter the activity of adrenal chromaffin cells. The effect of immune activation by lipopolysaccharide (LPS) on the regulation of tyrosine hydroxylase (TH) in the adrenal medulla in vivo was determined between 1 day and 6 months after LPS injection. The plasma levels of eleven cytokines were reduced 1 day after LPS injection, whereas the level for interleukin-10 was increased. The levels of all cytokines remained at control levels until 6 months when the levels of interleukin-6 and -4 were increased. One day after LPS injection, there was a decrease in TH-specific activity that may be due to decreased phosphorylation of serine 31 and 40. This decreased phosphorylation of serine 31 and 40 may be due to an increased activation of the protein phosphatase PP2A. One week after LPS injection, there was increased TH protein and increased phosphorylation of serine 40 that this was not accompanied by an increase in TH-specific activity. All TH parameters measured returned to basal levels between 1 month and 3 months. Six months after injection there was an increase in TH protein. This was associated with increased levels of the extracellular regulated kinase isoforms 1 and 2. This work shows that a single inflammatory event has the capacity to generate both short-term and long-term changes in TH regulation in the adrenal medulla of the adult animal. J. Cell. Biochem. 118: 2096-2107, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adrenal Medulla/drug effects , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Tyrosine 3-Monooxygenase/genetics , Adrenal Medulla/immunology , Adrenal Medulla/pathology , Animals , Body Weight/drug effects , Cytokines/genetics , Cytokines/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Phosphorylation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/immunology , Rats , Rats, Sprague-Dawley , Signal Transduction , Tyrosine 3-Monooxygenase/immunology
13.
Brain Behav Immun ; 60: 117-125, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27717686

ABSTRACT

Exposure to psychological stress is known to seriously disrupt the operation of the substantia nigra (SN) and may in fact initiate the loss of dopaminergic neurons within the SN. In this study, we aimed to investigate how chronic stress modified the SN in adult male mice. Using a paradigm of repeated restraint stress (an average of 20h per week for 6weeks), we examined changes within the SN using western blotting and immunohistochemistry. We demonstrated that chronic stress was associated with a clear loss of dopaminergic neurons within the SN. The loss of dopaminergic neurons was accompanied by higher levels of oxidative stress damage, indexed by levels of protein carbonylation and strong suppression of both microglial and astrocytic responses. In addition, we demonstrated for the first time, that chronic stress alone enhanced the aggregation of α-synuclein into the insoluble protein fraction. These results indicate that chronic stress triggered loss of dopaminergic neurons by increasing oxidative stress, suppressing glial neuroprotective functions and enhancing the aggregation of the neurotoxic protein, α-synuclein. Collectively, these results reinforce the negative effects of chronic stress on the viability of dopaminergic cells within the SN.


Subject(s)
Astrocytes/metabolism , Dopaminergic Neurons/metabolism , Microglia/metabolism , Neuroglia/metabolism , Substantia Nigra/metabolism , Animals , Male , Mice, Inbred C57BL , Oxidative Stress/physiology , Stress, Physiological/physiology , alpha-Synuclein/metabolism
14.
Mol Neurobiol ; 54(9): 6903-6916, 2017 11.
Article in English | MEDLINE | ID: mdl-27771902

ABSTRACT

Human neuroblastoma SH-SY5Y cells have been used as an in vitro model for neurodegenerative disorders such as Parkinson's disease and can be induced to a mature neuronal phenotype through retinoic acid (RA) differentiation. However, mechanisms of RA-induced differentiation remain unclear. Here, we investigate the role of reactive species (RS) on SH-SY5Y neuroblastoma cells under RA differentiation, using the antioxidant Trolox® as co-treatment. We found that RA treatment for 7 days reduced the cell number and proliferative capacity and induced the expression of adult catecholaminergic/neuronal markers such as tyrosine hydroxylase (TH), ß-III tubulin, and enolase-2. Evaluation of intracellular RS production by DCFH oxidation assay and quantification of cell non-enzymatic antioxidant activity by TRAP demonstrated that RA increases RS production. Furthermore, mitochondrial NADH oxidation showed to be inhibited under differentiation with RA. Cells subjected to co-treatment with antioxidant Trolox® demonstrated a remaining proliferative capacity and a decrease in the pro-oxidant state and RS production. Besides, antioxidant treatment restores the mitochondrial NADH oxidation. Importantly, Trolox® co-treatment inhibited the appearance of morphological characteristics such as neurite extension and branching, and decreased the expression of TH, ß-III tubulin, and enolase-2 after a seven-day differentiation with RA, indicating that RS production is a necessary step in this process. Trolox® also inhibited the phosphorylation of Akt and ERK1/2, which are involved in differentiation and survival, respectively, of these cells. Altogether, these data indicate the presence of a redox-dependent mechanism in SH-SY5Y RA-differentiation process and can be a useful insight to improve understanding of neuronal differentiation signaling.


Subject(s)
Biomarkers/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Neurons/cytology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology , Up-Regulation/drug effects , Antioxidants/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Phenotype , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
15.
Eur J Pharmacol ; 784: 147-54, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27181066

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) regulates synaptic protein synthesis and therefore synaptic function and plasticity. A role for mTORC1 has recently been demonstrated for addiction-related behaviors. For example, central or intra-accumbal injections of the mTORC1 inhibitor rapamycin attenuates several indices of cocaine-seeking including progressive ratio (PR) responding and reinstatement. These behavioral effects are associated with decreased mTORC1 activity and synaptic protein translation in the nucleus accumbens (NAC) and point to a possible therapeutic role for rapamycin in the treatment of addiction. Currently, it is unclear whether similar behavioral and biochemical effects can be achieved by administering rapamycin systemically, which represents a more clinically-appropriate route of administration. Here, we assessed the effects of repeated, systemic administration of rapamycin (10mg/kg, i.p.) on PR responding for cocaine. We also assessed whether systemic rapamycin was associated with changes in measures of mTORC1 activity and GluA1 expression in the ventral and dorsal striatum. We report that systemic rapamycin treatment reduced PR breakpoints to levels comparable to intra-NAC rapamycin. Systemic rapamycin treatment also reduced phosphorylated p70S6K and GluA1 AMPARs within the NAC but not dorsal striatum. Thus, systemic administration of rapamycin is as effective at reducing drug seeking behavior and measures of mTORC1 activity compared to direct accumbal application and may therefore represent a possible therapeutic option in the treatment of addiction. Possible caveats of this treatment approach are discussed.


Subject(s)
Cocaine/pharmacology , Gene Expression Regulation/drug effects , Motivation/drug effects , Receptors, AMPA/metabolism , Sirolimus/pharmacology , Ventral Striatum/drug effects , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Rats , Rats, Sprague-Dawley , Self Administration/psychology , TOR Serine-Threonine Kinases/metabolism , Ventral Striatum/metabolism , Ventral Striatum/physiology
16.
Metallomics ; 8(6): 597-604, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26790482

ABSTRACT

Manganese (Mn) is an essential trace element required for a range of physiological processes, but Mn can also be neurotoxic especially during development. Excess levels of Mn accumulate preferentially in the striatum and can induce a syndrome called manganism, characterized by an initial stage of psychiatric disorder followed by motor impairment. In the present study, we investigated the effects of Mn exposure on the developing dopaminergic system, specifically tyrosine hydroxylase (TH) protein and phosphorylation levels in the striatum of rats. Neonatal rats were exposed to Mn intraperitoneally (ip) from post-natal day 8 up to day 12 (PND8-12). Striatal tissue was analysed on PND14 or PND70, to detect either short-term or long-term effects induced by Mn exposure. There was a dose dependent increase in TH protein levels in the striatum at PND14, reaching significance at 20 mg kg(-1) Mn, and this correlated with an increase in TH phosphorylation at serines 40, 31 and 19. However, in the striatum at PND70, a time by which Mn levels were no longer elevated, there was a dose dependent decrease in TH protein levels, reaching significance at 20 mg kg(-1) Mn, and this correlated with TH phosphorylation at Ser40 and Ser19. There was however a significant increase in phosphorylation of TH at serine 31 at 20 mg kg(-1) Mn, which did not correlate with TH protein levels. Taken together our findings suggest that neonatal Mn exposure can have both short-term and long-term effects on the regulation of TH in the striatal dopaminergic system.


Subject(s)
Corpus Striatum/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Manganese/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Animals , Animals, Newborn , Corpus Striatum/drug effects , Male , Phosphorylation/drug effects , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/genetics
17.
Neuropsychopharmacology ; 39(7): 1694-702, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24469593

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is necessary for synaptic plasticity, as it is critically involved in the translation of synaptic transmission-related proteins, such as Ca(2+)/Calmodulin-dependent kinase II alpha (CAMKIIα) and AMPA receptor subunits (GluAs). Although recent studies have implicated mTORC1 signaling in drug-motivated behavior, the ineffectiveness of rapamycin, an mTORC1 inhibitor, in suppressing cocaine self-administration has raised questions regarding the specific role of mTORC1 in drug-related behaviors. Here, we examined mTORC1's role in three drug-related behaviors: cocaine taking, withdrawal, and reinstatement of cocaine seeking, by measuring indices of mTORC1 activity and assessing the effect of intra-cerebroventricular rapamycin on these behaviors in rats. We found that withdrawal from cocaine self-administration increased indices of mTORC1 activity in the nucleus accumbens (NAC). Intra-cerebroventricular rapamycin attenuated progressive ratio (PR) break points and reduced phospho-p70 ribosomal S6 kinase, GluA1 AMPAR, and CAMKIIα levels in the NAC shell (NACsh) and core (NACc). In a subsequent study, we treated rats with intra-NACsh infusions of rapamycin (2.5 µg/side/day for 5 days) during cocaine self-administration and then tracked the expression of addiction-relevant behaviors through to withdrawal and extinction. Rapamycin reduced drug seeking in signaled non-drug-available periods, PR responding, and cue-induced reinstatement, with these effects linked to reduced mTORC1 activity, total CAMKIIα, and GluA1 AMPAR levels in the NACsh. Together, these data highlight a role for mTORC1 in the neural processes that control the expression and maintenance of drug reward, including protracted relapse vulnerability. These effects appear to involve a role for mTORC1 in the regulation of GluA1 AMPARs and CAMKIIα in the NACsh.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Drug-Seeking Behavior/drug effects , Multiprotein Complexes/antagonists & inhibitors , Nucleus Accumbens/drug effects , Receptors, AMPA/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cocaine/adverse effects , Cocaine-Related Disorders/prevention & control , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/adverse effects , Gene Expression Regulation/drug effects , Mechanistic Target of Rapamycin Complex 1 , Motor Activity , Multiprotein Complexes/metabolism , Rats , Recurrence , Reinforcement, Psychology , Self Administration , TOR Serine-Threonine Kinases/metabolism
18.
Cell Signal ; 26(4): 748-56, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24407174

ABSTRACT

Calcium/calmodulin-stimulated protein kinase II (CaMKII) is a multi-functional serine/threonine protein kinase that controls a range of cellular functions, including proliferation. The biological properties of CaMKII are regulated by multi-site phosphorylation and targeting via interactions with specific proteins. To investigate the role specific CaMKII phosphorylation sites play in controlling cell proliferation and cell cycle progression, we examined phosphorylation of CaMKII at two sites (T253 and T286) at various stages of the cell cycle, and also examined the effects of overexpression of wild-type (WT), T286D phosphomimic, T253D phosphomimic and T253V phosphonull forms of CaMKIIα in MDA-MB-231 breast cancer and SHSY5Y neuroblastoma cells on cellular proliferation and cell cycle progression. We demonstrate herein that whilst there is no change in total CaMKII expression or T286 phosphorylation throughout the cell cycle, a marked dephosphorylation of CaMKII at T253 occurs during the G2 and/or M phases. Additionally, we show by molecular inhibition, as well as pharmacological activation, that protein phosphatase 2A (PP2A) is the phosphatase responsible for this dephosphorylation. Furthermore, we show that inducible overexpression of WT, T286D and T253V forms of CaMKIIα in MDA-MB-231 and SHSY5Y cells increases cellular proliferation, with no alteration in cell cycle profiles. By contrast, overexpression of a T253D phosphomimic form of CaMKIIα significantly decreases proliferation, and cells accumulate in mitosis, specifically in metaphase. Taken together, these results strongly suggest that the dephosphorylation of CaMKII at T253 is involved in controlling the cell cycle, specifically the metaphase-anaphase transition.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Amino Acid Substitution , Anaphase , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Metaphase , Mitosis , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , RNA Interference , RNA, Small Interfering/metabolism
19.
J Neurochem ; 128(6): 829-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24117434

ABSTRACT

Müller cells constitute the main glial cell type in the retina where it interacts with virtually all cells displaying relevant functions to retinal physiology. Under appropriate stimuli, Müller cells may undergo dedifferentiation, being able to generate other neural cell types. Here, we show that purified mouse Müller cells in culture express a group of proteins related to the dopaminergic phenotype, including the nuclear receptor-related 1 protein, required for dopaminergic differentiation, as well the enzyme tyrosine hydroxylase. These dopaminergic components are active, since Müller cells are able to synthesize and release dopamine to the extracellular medium. Moreover, Müller-derived tyrosine hydroxylase can be regulated, increasing its activity because of phosphorylation of serine residues in response to agents that increase intracellular cAMP levels. These observations were extended to glial cells obtained from adult monkey retinas with essentially the same results. To address the potential use of dopaminergic Müller cells as a source of dopamine in cell therapy procedures, we used a mouse model of Parkinson's disease, in which mouse Müller cells with the dopaminergic phenotype were transplanted into the striatum of hemi-parkinsonian mice generated by unilateral injection of 6-hydroxydopamine. These cells fully decreased the apomorphine-induced rotational behavior and restored motor functions in these animals, as measured by the rotarod and the forelimb-use asymmetry (cylinder) tests. The data indicate local restoration of dopaminergic signaling in hemi-parkinsonian mice confirmed by measurement of striatal dopamine after Müller cell grafting.


Subject(s)
Dopaminergic Neurons/transplantation , Ependymoglial Cells/transplantation , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , Animals , Cebus , Cell Differentiation/physiology , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/physiology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinsonian Disorders/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Recovery of Function/physiology , Tyrosine 3-Monooxygenase/metabolism
20.
J Neurochem ; 128(4): 547-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24117713

ABSTRACT

Stress activates selected neuronal systems in the brain and this leads to activation of a range of effector systems. Our aim was to investigate some of the relationships between these systems under basal conditions and over a 40-min period in response to footshock stress. Specifically, we investigated catecholaminergic neurons in the locus coeruleus (LC), ventral tegmental area and medial prefrontal cortex (mPFC) in the brain, by measuring tyrosine hydroxylase (TH) protein, TH phosphorylation and TH activation. We also measured the effector responses by measuring plasma adrenocorticotrophic hormone, corticosterone, glucose and body temperature as well as activation of adrenal medulla protein kinases, TH protein, TH phosphorylation and TH activation. The LC, ventral tegmental area and adrenal medulla all had higher basal levels of Ser19 phosphorylation and lower basal levels of Ser31 phosphorylation than the mPFC, presumably because of their cell body versus nerve terminal location, while the adrenal medulla had the highest basal levels of Ser40 phosphorylation. Ser31 phosphorylation was increased in the LC at 20 and 40 min and in the mPFC at 40 min; TH activity was increased at 40 min in both tissues. There were significant increases in body temperature between 10 and 40 min, as well as increases in plasma adrenocorticotropic hormone at 20 min and corticosterone and glucose at 20 and 40 min. The adrenal medulla extracellular signal-regulated kinase 2 was increased between 10 and 40 min and Ser31 phosphorylation was increased at 20 min and 40 min. Protein kinase A and Ser40 phosphorylation were increased only at 40 min. TH activity was increased between 20 and 40 min. TH protein and Ser19 phosphorylation levels were not altered in any of the brain regions or adrenal medulla over the first 40 min. These findings indicate that acute footshock stress leads to activation of TH in the LC, pre-synaptic terminals in the mPFC and adrenal medullary chromaffin cells, as well as changes in activity of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Adrenal Medulla/pathology , Brain/pathology , Electroshock , Stress, Psychological/pathology , Tyrosine 3-Monooxygenase/metabolism , Adrenal Medulla/enzymology , Adrenocorticotropic Hormone/blood , Animals , Blood Glucose/analysis , Blotting, Western , Body Temperature , Brain/enzymology , Corticosterone/blood , Enzyme Activation/physiology , Locus Coeruleus/metabolism , Male , Phosphorylation , Prefrontal Cortex/metabolism , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/physiology , Ventral Tegmental Area/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...