Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cells ; 11(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35883654

ABSTRACT

Autism Spectrum Disorder (ASD) is a common pediatric neurobiological disorder with up to 80% of genetic etiologies. Systems biology approaches may make it possible to test novel therapeutic strategies targeting molecular pathways to alleviate ASD symptoms. A clinical database of autism subjects was queried for individuals with a copy number variation (CNV) on microarray, Vineland, and Parent Concern Questionnaire scores. Pathway analyses of genes from pathogenic CNVs yielded 659 genes whose protein-protein interactions and mRNA expression mapped 121 genes with maximal antenatal expression in 12 brain regions. A Research Domain Criteria (RDoC)-derived neural circuits map revealed significant differences in anxiety, motor, and activities of daily living skills scores between altered CNV genes and normal microarrays subjects, involving Positive Valence (reward), Cognition (IQ), and Social Processes. Vascular signaling was identified as a biological process that may influence these neural circuits. Neuroinflammation, microglial activation, iNOS and 3-nitrotyrosine increase in the brain of Semaphorin 3F- Neuropilin 2 (Sema 3F-NRP2) KO, an ASD mouse model, agree with previous reports in the brain of ASD individuals. Signs of platelet deposition, activation, release of serotonin, and albumin leakage in ASD-relevant brain regions suggest possible blood brain barrier (BBB) deficits. Disruption of neurovascular signaling and BBB with neuroinflammation may mediate causative pathophysiology in some ASD subgroups. Although preliminary, these data demonstrate the potential for developing novel therapeutic strategies based on clinically derived data, genomics, cognitive neuroscience, and basic neuroscience methods.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Activities of Daily Living , Animals , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Blood-Brain Barrier/metabolism , DNA Copy Number Variations , Female , Humans , Mice , Pilot Projects , Pregnancy
2.
J Appl Physiol (1985) ; 131(5): 1552-1564, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34590907

ABSTRACT

Microvascular dilation, important for peripheral tissue glucose distribution, also modulates alveolar perfusion and is inhibited by loss of bioavailable nitric oxide (NO) in diabetes mellitus (DM). We hypothesized that DM-induced oxidative stress decreases bioavailable NO and pulmonary precapillary arteriolar diameter, causing endothelial injury. We examined subpleural pulmonary arterioles after acute NO synthase (NOS) inhibition with NG-nitro-l-arginine methyl ester (l-NAME) in streptozotocin (STZ)- and saline (CTRL)-treated C57BL/6J mice. Microvascular changes were assessed by intravital microscopy in the right lung of anesthetized mice with open chest and ventilated lungs. Arteriolar tone in pulmonary arterioles (27.2-48.7 µm diameter) increased in CTRL mice (18.0 ± 11% constriction, P = 0.034, n = 5) but decreased in STZ mice (13.6 ± 7.5% dilation, P = 0.009, n = 5) after l-NAME. Lung tissue dihydroethidium (DHE) fluorescence (superoxide), inducible NOS expression, and protein nitrosylation (3-nitrotyrosine) increased in STZ mice and correlated with increased glucose levels (103.8 ± 8.8 mg/dL). Fluorescently labeled fibrinogen administration and fibrinogen immunostaining showed fibrinogen adhesion, indicating endothelial injury in STZ mice. In CTRL mice, vasoconstriction to l-NAME was likely due to the loss of bioavailable NO. Vasodilation in STZ mice may be due to decreased formation of a vasoconstrictor or emergence of a vasodilator. These findings provide novel evidence that DM targets the pulmonary microcirculation and that decreased NO bioavailability and increased precapillary arteriolar tone could potentially lead to ventilation-perfusion abnormalities, exacerbating systemic DM complications.NEW & NOTEWORTHY Diabetes pulmonary and microvascular consequences are well recognized but have not been characterized. We assessed lung microvascular changes in a live anesthetized mouse model of type 1 diabetes, using a novel intravital microscopy technique. Our results show new evidence that a diabetes-induced decrease in lung nitric oxide bioavailability underlies oxidative damage, enhanced platelet activation, and endothelial injury causing pulmonary microvascular dysfunction and altered vasoreactivity. These findings could provide novel strategies to prevent or reverse diabetes systemic consequences.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Animals , Arterioles , Lung , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide , Oxidative Stress , Vasodilation
3.
J Neurochem ; 159(1): 15-28, 2021 10.
Article in English | MEDLINE | ID: mdl-34169527

ABSTRACT

Autism Spectrum Disorder (ASD) is a neurodevelopmental disease originating from combined genetic and environmental factors. Post-mortem human studies and some animal ASD models have shown brain neuroinflammation, oxidative stress, and changes in blood-brain barrier (BBB) integrity. However, the signaling pathways leading to these inflammatory findings and vascular alterations are currently unclear. The BBB plays a critical role in controlling brain homeostasis and immune response. Its dysfunction can result from developmental genetic abnormalities or neuroinflammatory processes. In this review, we explore the role of the Sonic Hedgehog/Wingless-related integration site (Shh/Wnt) pathways in neurodevelopment, neuroinflammation, and BBB development. The balance between Wnt-ß-catenin and Shh pathways controls angiogenesis, barriergenesis, neurodevelopment, central nervous system (CNS) morphogenesis, and neuronal guidance. These interactions are critical to maintain BBB function in the mature CNS to prevent the influx of pathogens and inflammatory cells. Genetic mutations of key components of these pathways have been identified in ASD patients and animal models, which correlate with the severity of ASD symptoms. Disruption of the Shh/Wnt crosstalk may therefore compromise BBB development and function. In turn, impaired Shh signaling and glial activation may cause neuroinflammation that could disrupt the BBB. Elucidating how ASD-related mutations of Shh/Wnt signaling could cause BBB leaks and neuroinflammation will contribute to our understanding of the role of their interactions in ASD pathophysiology. These observations may provide novel targeted therapeutic strategies to prevent or alleviate ASD symptoms while preserving normal developmental processes. Cover Image for this issue: https://doi.org/10.1111/jnc.15081.


Subject(s)
Autism Spectrum Disorder/metabolism , Blood-Brain Barrier/metabolism , Hedgehog Proteins/metabolism , Neurovascular Coupling/physiology , Wnt Signaling Pathway/physiology , Animals , Autism Spectrum Disorder/genetics , Hedgehog Proteins/genetics , Humans , Mutation/physiology , Tight Junctions/genetics , Tight Junctions/metabolism
4.
J Cell Mol Med ; 25(14): 6828-6840, 2021 07.
Article in English | MEDLINE | ID: mdl-34053181

ABSTRACT

To efficiently prevent diabetic cardiomyopathy (DCM), we have explored and confirmed that metallothionein (MT) prevents DCM by attenuating oxidative stress, and increasing expression of proteins associated with glucose metabolism. To determine whether Akt2 expression is critical to MT prevention of DCM, mice with either global Akt2 gene deletion (Akt2-KO), or cardiomyocyte-specific overexpressing MT gene (MT-TG) or both combined (MT-TG/Akt2-KO) were used. Akt2-KO mice exhibited symptoms of DCM (cardiac remodelling and dysfunction), and reduced expression of glycogen and glucose metabolism-related proteins, despite an increase in total Akt (t-Akt) phosphorylation. Cardiac MT overexpression in MT-TG/Akt2-KO mice prevented DCM and restored glucose metabolism-related proteins expression and baseline t-Akt phosphorylation. Furthermore, phosphorylation of ERK1/2 increased in the heart of MT-TG/Akt2-KO mice, compared with Akt2-KO mice. As ERK1/2 has been implicated in the regulation of glucose transport and metabolism this increase could potentially underlie MT protective effect in MT-TG/Akt2-KO mice. Therefore, these results show that although our previous work has shown that MT preserving Akt2 activity is sufficient to prevent DCM, in the absence of Akt2 MT may stimulate alternative or downstream pathways protecting from DCM in a type 2 model of diabetes, and that this protection may be associated with the ERK activation pathway.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Metallothionein/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Diabetic Cardiomyopathies/genetics , Female , Glucose/metabolism , Humans , Male , Metallothionein/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/metabolism , Proto-Oncogene Proteins c-akt/deficiency , Transgenes , Up-Regulation
5.
Toxicol Rep ; 7: 1164-1169, 2020.
Article in English | MEDLINE | ID: mdl-32983904

ABSTRACT

Analyses of human cohort data support the roles of cadmium and obesity in the development of several neurocognitive disorders. To explore the effects of cadmium exposure in the brain, mice were subjected to whole life oral cadmium exposure. There were significant increases in cadmium levels with female animals accumulating more metal than males (p < 0.001). Both genders fed a high fat diet showed significant increases in cadmium levels compared to low fat diet fed mice (p < 0.001). Cadmium and high fat diet significantly affected the levels of several essential metals, including magnesium, potassium, chromium, iron, cobalt, copper, zinc and selenium. Additionally, these treatments resulted in increased superoxide levels within the cortex, amygdala and hippocampus. These findings support a model where cadmium and high fat diet affect the levels of redox-active, essential metal homeostasis. This phenomenon may contribute to the underlying mechanism(s) responsible for the development of neurocognitive disorders.

6.
Inflamm Res ; 69(1): 15-26, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31707449

ABSTRACT

BACKGROUND: Intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), is prevalent in older adults and associated with inflammation. We previously showed that IH induces renal fibrosis and cardiomyopathy and hypothesized that lung inflammatory changes may underlie deficits in pulmonary function in OSA. METHODS: Pulmonary inflammatory and oxidative markers were assessed in metallothionein KO (MT-KO) mice and WT 129S1 controls exposed to IH or to normoxia for 8 weeks. RESULTS: MT expression increased at 3 days in WT, falling back at 1 week. Pro-fibrotic markers CTGF and PAI-1 were unchanged in WT, but increased at 3 or 8 weeks, with enhanced Sirius Red staining at 8 weeks, in IH-exposed MT-KO. Cellular infiltration, TNF-α and IL-6 increased earlier in IH-exposed MT-KO than in WT. Oxidative markers, 3-nitrotyrosine and 4-hydroxynonenal increased in both but persisted in MT-KO. Antioxidant Nrf2, HO-1 and NQO1, increased at 3 days in WT mice and at 8 weeks IH in MT-KO. While early Nrf2 induction required MT, its later increase at 8 weeks in MT-KO was independent from MT. CONCLUSIONS: We conclude that early MT and antioxidant gene response protects from fibrotic changes in long-term IH-exposed mouse lung. Without this response, pulmonary fibrosis may develop with longer IH exposure.


Subject(s)
Hypoxia/metabolism , Lung Injury/metabolism , Metallothionein/metabolism , Animals , Hypoxia/complications , Hypoxia/pathology , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Metallothionein/genetics , Mice, Knockout , Oxidative Stress , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Biol Sci ; 15(1): 239-252, 2019.
Article in English | MEDLINE | ID: mdl-30662363

ABSTRACT

Hyperglycemia-induced renal fibrosis causes end-stage renal disease. Clopidogrel, a platelet inhibitor, is often administered to decrease cardiovascular events in diabetic patients. We investigated whether clopidogrel can reduce diabetes-induced renal fibrosis in a streptozotocin-induced type 1 diabetes murine model and fibronectin involvement in this protective response. Diabetic and age-matched controls were sacrificed three months after the onset of diabetes, and additional controls and diabetic animals were further treated with clopidogrel or vehicle for three months. Diabetes induced renal morphological changes and fibrosis after three months. Clopidogrel, administered during the last three months, significantly decreased blood glucose, collagen and fibronectin expression compared to vehicle-treated diabetic mice. Diabetes increased TGF-ß expression, inducing fibrosis via Smad-independent pathways, MAP kinases, and Akt activation at three months but returned to baseline at six months, whereas the expression of fibronectin and collagen remained elevated. Our results suggest that activation of TGF-ß, CTGF, and MAP kinases are early profibrotic signaling events, resulting in significant fibronectin accumulation at the early time point and returning to baseline at a later time point. Akt activation at the three-month time point may serve as an adaptive response in T1D. Mechanisms of clopidogrel therapeutic effect on the diabetic kidney remain to be investigated as this clinically approved compound could provide novel approaches to prevent diabetes-induced renal disease, therefore improving patients' survival.


Subject(s)
Clopidogrel/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Fibronectins/metabolism , Fibrosis/drug therapy , Fibrosis/etiology , Kidney Diseases/drug therapy , Animals , Blood Coagulation/drug effects , Blotting, Western , Clopidogrel/pharmacology , Fibrosis/metabolism , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Purinergic P2Y Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/therapeutic use
8.
Mol Neurobiol ; 56(8): 5520-5538, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30635860

ABSTRACT

Autism and epilepsy are diseases which have complex genetic inheritance. Genome-wide association and other genetic studies have implicated at least 500+ genes associated with the occurrence of autism spectrum disorders (ASD) including the human semaphorin 3F (Sema 3F) and neuropilin 2 (NRP2) genes. However, the genetic basis of the comorbid occurrence of autism and epilepsy is unknown. The aberrant development of GABAergic circuitry is a possible risk factor in autism and epilepsy. Molecular biological approaches were used to test the hypothesis that cell-specific genetic variation in mouse homologs affects the formation and function of GABAergic circuitry. The empirical analysis with mice homozygous null for one of these genes, Sema 3F, in GABAergic neurons substantiated these predictions. Notably, deletion of Sema 3F in interneurons but not excitatory neurons during early development decreased the number of interneurons/neurites and mRNAs for cell-specific GABAergic markers and increased epileptogenesis and autistic behaviors. Studies of interneuron cell-specific knockout of Sema 3F signaling suggest that deficient Sema 3F signaling may lead to neuroinflammation and oxidative stress. Further studies of mouse KO models of ASD genes such as Sema 3F or NRP2 may be informative to clinical phenotypes contributing to the pathogenesis in autism and epilepsy patients.


Subject(s)
Autistic Disorder/metabolism , Autistic Disorder/pathology , Behavior, Animal , GABAergic Neurons/metabolism , Interneurons/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Oxidative Stress , Animals , Biomarkers/metabolism , Cell Count , Epilepsy/metabolism , Epilepsy/pathology , Gene Deletion , Inflammation/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Reactive Oxygen Species/metabolism , Recombination, Genetic/genetics , Social Behavior
9.
Neuromolecular Med ; 20(2): 161-173, 2018 06.
Article in English | MEDLINE | ID: mdl-29691724

ABSTRACT

Autism spectrum disorders (ASD) are the most prevalent set of pediatric neurobiological disorders. The etiology of ASD has both genetic and environmental components including possible dysfunction of the immune system. The relationship of the immune system to aberrant neural circuitry output in the form of altered behaviors and communication characterized by ASD is unknown. Dysregulation of neurotrophins such as BDNF and their signaling pathways have been implicated in ASD. While abnormal cortical formation and autistic behaviors in mouse models of immune activation have been described, no one theory has been described to link activation of the immune system to specific brain signaling pathways aberrant in ASD. In this paper we explore the relationship between neurotrophin signaling, the immune system and ASD. To this effect we hypothesize that an interplay of dysregulated immune system, synaptogenic growth factors and their signaling pathways contribute to the development of ASD phenotypes.


Subject(s)
Autism Spectrum Disorder/therapy , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/immunology , Brain-Derived Neurotrophic Factor/immunology , Child , Gene-Environment Interaction , Humans , Immunization/adverse effects , Infections/immunology , Intercellular Signaling Peptides and Proteins/physiology , Microglia/immunology , Models, Immunological , Nerve Growth Factors/physiology , Neuroimmunomodulation , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/physiology , Wnt Signaling Pathway/immunology
10.
Toxicol Lett ; 279: 107-114, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28751209

ABSTRACT

Hypoxia is a primary factor in many pathological conditions. Hypoxic cell death is commonly attributed to metabolic failure and oxidative injury. cAMP-dependent protein kinase A (PKA) is activated in hypoxia and regulates multiple enzymes of the mitochondrial electron transport chain, thus may be implicated in cellular energy depletion and hypoxia-induced cell death. Wild type (WT) PC-12 cells and PKA activity-deficient 123.7 PC-12 cells were exposed to 3, 6, 12 and 24h hypoxia (0.1% or 5% O2). Hypoxia, at 24h 0.1% O2, induced cell death and increased reactive oxygen species (ROS) in WT PC-12 cells. Despite lower ATP levels in normoxic 123.7 cells than in WT cells, hypoxia only decreased ATP levels in WT cells. However, menadione-induced oxidative stress similarly affected both cell types. While mitochondrial COX IV expression remained consistently higher in 123.7 cells, hypoxia decreased COX IV expression in both cell types. N-acetyl cysteine antioxidant treatment blocked hypoxia-induced WT cell death without preventing ATP depletion. Transient PKA catα expression in 123.7 cells partially restored hypoxia-induced ROS but did not alter ATP levels or COX IV expression. We conclude that PKA signaling contributes to hypoxic injury, by regulating oxidative stress rather than by depleting ATP levels. Therapeutic strategies targeting PKA signaling may improve cellular adaptation and recovery in hypoxic pathologies.


Subject(s)
Adrenal Gland Neoplasms/enzymology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Neurons/enzymology , Oxidative Stress , Pheochromocytoma/enzymology , Reactive Oxygen Species/metabolism , Tumor Hypoxia , Adenosine Triphosphate/metabolism , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Animals , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Electron Transport Complex IV/metabolism , Energy Metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , PC12 Cells , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Rats , Signal Transduction , Time Factors , Transfection , Vitamin K 3/pharmacology
11.
Int J Mol Sci ; 17(11)2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27834824

ABSTRACT

Diabetes is strongly associated with systemic inflammation and oxidative stress, but its effect on pulmonary vascular disease and lung function has often been disregarded. Several studies identified restrictive lung disease and fibrotic changes in diabetic patients and in animal models of diabetes. While microvascular dysfunction is a well-known complication of diabetes, the mechanisms leading to diabetes-induced lung injury have largely been disregarded. We described the potential involvement of diabetes-induced platelet-endothelial interactions in perpetuating vascular inflammation and oxidative injury leading to fibrotic changes in the lung. Changes in nitric oxide synthase (NOS) activation and decreased NO bioavailability in the diabetic lung increase platelet activation and vascular injury and may account for platelet hyperreactivity reported in diabetic patients. Additionally, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has been reported to mediate pancreatic islet damage, and is implicated in the onset of diabetes, inflammation and vascular injury. Many growth factors and diabetes-induced agonists act via the JAK/STAT pathway. Other studies reported the contribution of the JAK/STAT pathway to the regulation of the pulmonary fibrotic process but the role of this pathway in the development of diabetic lung fibrosis has not been considered. These observations may open new therapeutic perspectives for modulating multiple pathways to mitigate diabetes onset or its pulmonary consequences.


Subject(s)
Blood Platelets/pathology , Diabetes Mellitus/pathology , Endothelial Cells/pathology , Lung/pathology , Peripheral Vascular Diseases/pathology , Pulmonary Fibrosis/pathology , Animals , Blood Platelets/metabolism , Cell Communication , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Inflammation , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Lung/blood supply , Lung/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Peripheral Vascular Diseases/genetics , Peripheral Vascular Diseases/metabolism , Platelet Activation , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
12.
Sci Rep ; 6: 34477, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708338

ABSTRACT

Misfolded alpha-synuclein (AS) and other neurodegenerative disorder proteins display prion-like transmission of protein aggregation. Factors responsible for the initiation of AS aggregation are unknown. To evaluate the role of amyloid proteins made by the microbiota we exposed aged rats and transgenic C. elegans to E. coli producing the extracellular bacterial amyloid protein curli. Rats exposed to curli-producing bacteria displayed increased neuronal AS deposition in both gut and brain and enhanced microgliosis and astrogliosis compared to rats exposed to either mutant bacteria unable to synthesize curli, or to vehicle alone. Animals exposed to curli producing bacteria also had more expression of TLR2, IL-6 and TNF in the brain than the other two groups. There were no differences among the rat groups in survival, body weight, inflammation in the mouth, retina, kidneys or gut epithelia, and circulating cytokine levels. AS-expressing C. elegans fed on curli-producing bacteria also had enhanced AS aggregation. These results suggest that bacterial amyloid functions as a trigger to initiate AS aggregation through cross-seeding and also primes responses of the innate immune system.


Subject(s)
Amyloid/pharmacology , Bacterial Proteins/pharmacology , Caenorhabditis elegans/metabolism , Escherichia coli Proteins/pharmacology , Escherichia coli , Protein Aggregation, Pathological/chemically induced , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Animals , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Rats , Rats, Inbred F344
13.
Physiol Rep ; 4(17)2016 09.
Article in English | MEDLINE | ID: mdl-27604401

ABSTRACT

Vascular dysfunction and decreased cerebral blood flow are linked to Alzheimer's disease (AD). Loss of endothelial nitric oxide (NO) and oxidative stress in human cerebrovascular endothelium increase expression of amyloid precursor protein (APP) and enhance production of the Aß peptide, suggesting that loss of endothelial NO contributes to AD pathology. We hypothesize that decreased systemic NO bioavailability in AD may also impact lung microcirculation and induce pulmonary endothelial dysfunction. The acute effect of NO synthase (NOS) inhibition on pulmonary arteriolar tone was assessed in a transgenic mouse model (TgAD) of AD (C57BL/6-Tg(Thy1-APPSwDutIowa)BWevn/Mmjax) and age-matched wild-type controls (C57BL/6J). Arteriolar diameters were measured before and after the administration of the NOS inhibitor, L-NAME Lung superoxide formation (DHE) and formation of nitrotyrosine (3-NT) were assessed as indicators of oxidative stress, inducible NOS (iNOS) and tumor necrosis factor alpha (TNF-α) expression as indicators of inflammation. Administration of L-NAME caused either significant pulmonary arteriolar constriction or no change from baseline tone in wild-type (WT) mice, and significant arteriolar dilation in TgAD mice. DHE, 3-NT, TNF-α, and iNOS expression were higher in TgAD lung tissue, compared to WT mice. These data suggest L-NAME could induce increased pulmonary arteriolar tone in WT mice from loss of bioavailable NO In contrast, NOS inhibition with L-NAME had a vasodilator effect in TgAD mice, potentially caused by decreased reactive nitrogen species formation, while significant oxidative stress and inflammation were present. We conclude that AD may increase pulmonary microvascular tone as a result of loss of bioavailable NO and increased oxidative stress. Our findings suggest that AD may have systemic microvascular implications beyond central neural control mechanisms.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Enzyme Inhibitors/administration & dosage , Lung/blood supply , Microcirculation/drug effects , NG-Nitroarginine Methyl Ester/administration & dosage , Oxidative Stress/drug effects , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Animals , Cerebrovascular Circulation/physiology , Disease Models, Animal , Endothelium/physiopathology , Enzyme Inhibitors/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Superoxides/metabolism
15.
PLoS One ; 9(7): e100230, 2014.
Article in English | MEDLINE | ID: mdl-25014412

ABSTRACT

Hypoxia alters cellular metabolism and although the effects of sustained hypoxia (SH) have been extensively studied, less is known about chronic intermittent hypoxia (IH), commonly associated with cardiovascular morbidity and stroke. We hypothesize that impaired glutamate homeostasis after chronic IH may underlie vulnerability to stroke-induced excitotoxicity. P16 organotypic hippocampal slices, cultured for 7 days were exposed for 7 days to IH (alternating 2 min 5% O2-15 min 21% O2), SH (5% O2) or RA (21% O2), then 3 glutamate challenges. The first and last exposures were intended as a metabolic stimulus (200 µM glutamate, 15 min); the second emulated excitotoxicity (10 mM glutamate, 10 min). GFAP, MAP2, and EAAT1, EAAT2 glutamate transporters expression were assessed after exposure to each hypoxic protocol. Additionally, cell viability was determined at baseline and after each glutamate challenge, in presence or absence of ceftriaxone that increases glutamate transporter expression. GFAP and MAP2 decreased after 7 days IH and SH. Long-term IH but not SH decreased EAAT1 and EAAT2. Excitotoxic glutamate challenge decreased cell viability and the following 200 µM exposure further increased cell death, particularly in IH-exposed slices. Ceftriaxone prevented glutamate transporter decrease and improved cell viability after IH and excitotoxicity. We conclude that IH is more detrimental to cell survival and glutamate homeostasis than SH. These findings suggest that impaired regulation of extracellular glutamate levels is implicated in the increased brain susceptibility to excitotoxic insult after long-term IH.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Ceftriaxone/pharmacology , Cell Hypoxia/physiology , Cell Survival/physiology , Animals , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/pharmacology , In Vitro Techniques , Rats , Rats, Sprague-Dawley
16.
Arch Physiol Biochem ; 118(2): 72-82, 2012 May.
Article in English | MEDLINE | ID: mdl-22439828

ABSTRACT

CONTEXT: Platelets have significant roles in initiating and mediating reduced alveolar blood flow, microvascular leak, and ventilation/perfusion mismatch caused by metabolic changes and altered signal transduction caused by ischemia-reperfusion. OBJECTIVE: This review focuses on platelet mechanisms of vascular dysfunction in the lung and presents a hypothesis for interplay between platelet activation, endothelial damage and fibrinogen. The purpose is to discuss current knowledge regarding mechanisms of platelet-mediated endothelial injury and implications for new strategies to treat vascular dysfunction associated with acute lung injury (ALI). METHODS: Literature from a number of fields was searched using Medline and Google Scholar. RESULTS: Activated platelets contribute to redox imbalance through reactive oxygen species production, pro-leak molecules such as PAF and serotonin, and recruitment of inflammatory cytokines and leukocytes to the damaged endothelium. CONCLUSION: Platelets are a critical component of pulmonary ALI, acting in conjunction with fibrinogen to mediate endothelial damage through multiple signal transduction pathways.


Subject(s)
Acute Lung Injury/complications , Blood Platelets/pathology , Vascular Diseases/complications , Vascular Diseases/physiopathology , Blood Platelets/metabolism , Blood Proteins/metabolism , Humans , Nitric Oxide/metabolism , Oxidative Stress , Vascular Diseases/blood , Vascular Diseases/metabolism
17.
Front Pharmacol ; 2: 96, 2011.
Article in English | MEDLINE | ID: mdl-22275901

ABSTRACT

Ever since it was shown for the first time that lactate can support neuronal function in vitro as a sole oxidative energy substrate, investigators in the field of neuroenergetics have been debating the role, if any, of this glycolytic product in cerebral energy metabolism. Our experiments employed the rat hippocampal slice preparation with electrophysiological and biochemical methodologies. The data generated by these experiments (a) support the hypothesis that lactate, not pyruvate, is the end-product of cerebral aerobic glycolysis; (b) indicate that lactate plays a major and crucial role in affording neural tissue to respond adequately to glutamate excitation and to recover unscathed post-excitation; (c) suggest that neural tissue activation is accompanied by aerobic lactate and NADH production, the latter being produced when the former is converted to pyruvate by mitochondrial lactate dehydrogenase (mLDH); (d) imply that NADH can be utilized as an endogenous scavenger of reactive oxygen species (ROS) to provide neuroprotection against ROS-induced neuronal damage.

18.
Neurochem Int ; 57(6): 655-61, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20708053

ABSTRACT

Identification of disease-associated proteins is critical for elucidating CNS disease mechanisms and elaborating novel treatment strategies. It requires post-mortem tissue analysis which can be significantly affected by the collection process, post-mortem intervals (PMIs), and storage conditions. To assess the effect of time and storage conditions on brain protein stability, SELDI-TOF-MS protein profiles were assessed in rat frontal cortex, caudate-putamen, hippocampus and medulla samples collected after various PMIs (0, 6, 12, 24, 48, and 72 h) at 4 °C or at room temperature (RT) storage. Regions of interest were isolated from cryosections (tissue apposition, TA), or micropunched from cryosections apposed on filter paper (paper apposition, PA), and applied onto an NP20 ProteinChip array. Protein alterations, while greater at RT than at 4 °C, were detected at 6h then differentially evolved in the various brain regions, with greater alterations in the caudate-putamen (60%) and the cortex (48%). Overall, our sensitive analytical method allowed unveiling of different patterns of protein susceptibility to PMI and to storage temperature in the various brain regions. Some protein peaks were altered in all brain regions and may potentially serve as markers of the PMI status of the brain, or for reference values when studying new proteins. Changes in disease-related proteins within post-mortem samples can be greatly affected by PMI and storage conditions, particularly when studying fragile and/or low abundant protein/peptides in tissues sampled from the caudate-putamen and neocortex.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Animals , Male , Proteomics , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Free Radic Biol Med ; 47(10): 1440-9, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19703551

ABSTRACT

Heat shock protein 90 (Hsp90) is a chaperone protein regulating PC-12 cell survival by binding and stabilizing Akt, Raf-1, and Cdc37. Hsp90 inhibitor geldanamycin (GA) cytotoxicity has been attributed to the disruption of Hsp90 binding, and the contribution of oxidative stress generated by its quinone group has not been studied in this context. Reactive oxygen species (ROS) and cell survival were assessed in PC-12 cells exposed to GA or menadione (MEN), and Akt, Raf-1, and Cdc37 expression and binding to Hsp90 were determined. GA disrupted Hsp90 binding and increased ROS production starting at 1 h, and cell death occurred at 6 h, inhibited by N-acetylcysteine (NAC) without preventing dissociation of proteins. At 24 h, NAC prevented cytotoxicity and Hsp90 complex disruption. However, MnTBAP antioxidant treatment failed to inhibit GA cytotoxicity, suggesting that NAC acts by restoring glutathione. In contrast, 24 h MEN treatment induced cytotoxicity without disrupting Hsp90 binding. GA and MEN decreased Hsp90-binding protein expression, and proteasomal inhibition prevented MEN-, but not GA-induced degradation. In conclusion, whereas MEN cytotoxicity is mediated by ROS and proteasomal degradation, GA-induced cytotoxicity requires ROS but induces Hsp90 complex dissociation and proteasome-independent protein degradation. These differences between MEN- and GA-induced cytotoxicity may allow more specific targeting of cancer cells.


Subject(s)
Benzoquinones/toxicity , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/toxicity , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/metabolism , PC12 Cells , Proteasome Endopeptidase Complex/metabolism , Rats , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Vitamin K 3/pharmacology
20.
Arch Physiol Biochem ; 115(1): 34-46, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19267281

ABSTRACT

Lung ischemia-reperfusion (IR) injury causes alveolar, epithelial and endothelial cell dysfunction which often results in decreased alveolar perfusion, characteristic of an acute respiratory distress syndrome. Nitric oxide (NO) from endothelium-derived NO synthase (eNOS) helps maintain a low pulmonary vascular resistance. Paradoxically, during acute lung injury, overproduction of NO via inducible NO synthase (iNOS) and oxidative stress lead to reactive oxygen and nitrogen species (ROS and RNS) formation and vascular dysfunction. RNS potentiate vascular and cellular injury by oxidation, by decreasing NO bioavailability, and by regulating NOS isoforms. RNS potentiate their own production by uncoupling NO production through eNOS by oxidation and disruption of Akt-mediated phosphorylation of eNOS. This review focuses on effects of NO which cause vascular dysfunction in the unique environment of the lung and presents a hypothesis for interplay between eNOS and iNOS activation with implications for development of new strategies to treat vascular dysfunction associated with IR.


Subject(s)
Lung , Nitric Oxide Synthase Type III/metabolism , Reperfusion Injury , Animals , HSP90 Heat-Shock Proteins/metabolism , Humans , Isoenzymes/metabolism , Lung/pathology , Lung/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Peroxynitrous Acid/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...