Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Blood Cells Mol Dis ; 86: 102493, 2021 02.
Article in English | MEDLINE | ID: mdl-32927249

ABSTRACT

Strokes are feared complications of sickle cell disease (SCD) and yield significant neurologic and neurocognitive deficits. However, even without detectable strokes, SCD patients have significant neurocognitive deficits in domains of learning and memory, processing speed and executive function. In these cases, mechanisms unrelated to major cerebrovascular abnormalities likely underlie these deficits. While oxidative stress and stress-related signaling pathways play a role in SCD pathophysiology, their role in cerebral injury remains unknown. We have shown that Townes and BERK SCD mice, while not having strokes, recapitulate neurocognitive deficits reported in humans. We hypothesized that cognitive deficits in SCD mice are associated with cerebral oxidative stress. We showed that SCD mice have increased levels of reactive oxygen species, protein carbonylation, and lipid peroxidation in hippocampus and cortex, thus suggesting increased cerebral oxidative stress. Further, cerebral oxidative stress was associated with caspase-3 activity alterations and vascular endothelial abnormalities, white matter changes, and disruption of the blood brain barrier, similar to those reported after ischemic/oxidative injury. Additionally, after repeated hypoxia/reoxygenation exposure, homozygous Townes had enhanced microglia activation. Our findings indicate that oxidative stress and stress-induced tissue damage is increased in susceptible brain regions, which may, in turn, contribute to neurocognitive deficits in SCD mice.


Subject(s)
Anemia, Sickle Cell/pathology , Endothelial Cells/pathology , Oxidative Stress , White Matter/pathology , Anemia, Sickle Cell/metabolism , Animals , Brain/metabolism , Brain/pathology , Cognition , Endothelial Cells/metabolism , Female , Humans , Male , Mice , White Matter/metabolism
2.
Haematologica ; 103(5): 787-798, 2018 05.
Article in English | MEDLINE | ID: mdl-29519868

ABSTRACT

Sickle cell disease patients are at increased risk of developing a chronic kidney disease. Endothelial dysfunction and inflammation associated with hemolysis lead to vasculopathy and contribute to the development of renal disease. Here we used a Townes sickle cell disease mouse model to examine renal endothelial injury. Renal disease in Townes mice was associated with glomerular hypertrophy, capillary dilation and congestion, and significant endothelial injury. We also detected substantial renal macrophage infiltration, and accumulation of macrophage stimulating protein 1 in glomerular capillary. Treatment of human cultured macrophages with hemin or red blood cell lysates significantly increased expression of macrophage membrane-associated protease that might cleave and activate circulating macrophage stimulating protein 1 precursor. Macrophage stimulating protein 1 binds to and activates RON kinase, a cell surface receptor tyrosine kinase. In cultured human renal glomerular endothelial cells, macrophage stimulating protein 1 induced RON downstream signaling, resulting in increased phosphorylation of ERK and AKT kinases, expression of Von Willebrand factor, increased cell motility, and re-organization of F-actin. Specificity of macrophage stimulating protein 1 function was confirmed by treatment with RON kinase inhibitor BMS-777607 that significantly reduced downstream signaling. Moreover, treatment of sickle cell mice with BMS-777607 significantly reduced glomerular hypertrophy, capillary dilation and congestion, and endothelial injury. Taken together, our findings demonstrated that RON kinase is involved in the induction of renal endothelial injury in sickle cell mice. Inhibition of RON kinase activation may provide a novel approach for prevention of the development of renal disease in sickle cell disease.


Subject(s)
Aminopyridines/pharmacology , Anemia, Sickle Cell/physiopathology , Endothelium, Vascular/drug effects , Kidney/drug effects , Macrophages/drug effects , Pyridones/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Humans , Kidney/injuries , Kidney/pathology , Macrophages/pathology , Mice
3.
Free Radic Biol Med ; 108: 533-541, 2017 07.
Article in English | MEDLINE | ID: mdl-28416347

ABSTRACT

The biology of the inorganic anion nitrite is linked to nitric oxide (NO) as nitrite can be reduced to NO and mediate its biological activities. Thus, studies of nitrite biology require sensitive and selective chemical assays. The acetic and ascorbic acids method is selective for nitrite and measures it in biological matrices. However, one of the pitfalls of nitrite measurements is its ubiquitous presence in sample collection tubes. Here, we showed high levels of nitrite in collection tubes containing EDTA, sodium citrate or sodium heparin and smaller amounts in tubes containing lithium heparin or serum clot activator. We also showed the presence of nitrite in colloid and crystalloid solutions frequently administered to patients and found variable levels of nitrite in 5% albumin, 0.9% sodium chloride, lactated ringer's, and dextrose-plus-sodium chloride solutions. These levels of nitrite varied across lots and manufacturers of the same type of fluid. Because these fluids are administered intravenously to patients (including those in shock), sometimes in large volumes (liters), it is possible that infusions of these nitrite-containing fluids may have clinical implications. A protocol for blood collection free of nitrite contamination was developed and used to examine nitrite levels in whole blood, red blood cells, plasma and urine from normal volunteers. Nitrite measurements were reproducible, had minimal variability, and did not indicate sex-differences. These findings validated a method and protocol for selective nitrite assay in biological fluids free of nitrite contamination which can be applied for study of diseases where dysfunctional NO signaling has been implicated.


Subject(s)
Blood Specimen Collection/methods , Blood Transfusion , Isotonic Solutions/chemistry , Nitric Oxide/chemistry , Nitrites/chemistry , Product Packaging , Administration, Intravenous , Citrates/chemistry , Crystalloid Solutions , Edetic Acid/chemistry , Heparin/chemistry , Humans , Isotonic Solutions/therapeutic use , Nitric Oxide/metabolism , Reproducibility of Results , Ringer's Lactate , Sensitivity and Specificity , Sodium Chloride/chemistry , Sodium Citrate
5.
Neuropharmacology ; 111: 323-334, 2016 12.
Article in English | MEDLINE | ID: mdl-27638450

ABSTRACT

Caretakers and clinicians alike have long recognized that individuals with autism spectrum disorder (ASD) can have altered sensory processing, which can contribute to its core symptoms. However, the pathobiology of sensory alterations in ASD is poorly understood. Here we examined nocifensive behavior in ASD mouse models, the BTBR T+Itpr3tf/J (BTBR) and the fragile-X mental retardation-1 knockout (Fmr1-KO) mice. We also examined the effects of nicotine on nocifensive behavior given that nicotine, a nicotinic cholinergic receptor (nAChR) agonist that has antinociceptive effects, was shown to improve social deficits and decrease repetitive behaviors in BTBR mice. Compared to respective controls, both BTBR and Fmr1-KO had hyporesponsiveness to noxious thermal stimuli and electrical stimulation of C-sensory fibers, normal responsiveness to electrical stimulation of Aß- and Aδ-fiber, and hyperresponsiveness to visceral pain after acetic acid intraperitoneal injection. In BTBR, nicotine at lower doses increased, whereas at higher doses, it decreased hotplate latency compared to vehicle. In a significantly different effect pattern, in control mice, nicotine had antinociceptive effects to noxious heat only at the high dose. Interestingly, these nocifensive behavior alterations and differential responses to nicotine antinociceptive effects in BTBR mice were associated with significant downregulation of α3, α4, α5, α7, ß2, ß3, and ß4 nAChR subunits in several cerebral regions both, during embryonic development and adulthood. Taken together, these findings further implicate nAChRs in behaviors alterations in the BTBR model and lend support to the hypothesis that nAChRs may be a target for treatment of behavior deficits and sensory dysfunction in ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/psychology , Avoidance Learning/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nociception/physiology , Receptors, Nicotinic/metabolism , Animals , Brain/embryology , Brain/metabolism , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociception/drug effects , Protein Subunits/metabolism
6.
Neurobiol Dis ; 85: 60-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26462816

ABSTRACT

Strokes are perhaps the most serious complications of sickle cell disease (SCD) and by the fifth decade occur in approximately 25% of patients. While most patients do not develop strokes, mounting evidence indicates that even without brain abnormalities on imaging studies, SCD patients can present profound neurocognitive dysfunction. We sought to evaluate the neurocognitive behavior profile of humanized SCD mice (Townes, BERK) and to identify hematologic and neuropathologic abnormalities associated with the behavioral alterations observed in these mice. Heterozygous and homozygous Townes mice displayed severe cognitive deficits shown by significant delays in spatial learning compared to controls. Homozygous Townes also had increased depression- and anxiety-like behaviors as well as reduced performance on voluntary wheel running compared to controls. Behavior deficits observed in Townes were also seen in BERKs. Interestingly, most deficits in homozygotes were observed in older mice and were associated with worsening anemia. Further, neuropathologic abnormalities including the presence of large bands of dark/pyknotic (shrunken) neurons in CA1 and CA3 fields of hippocampus and evidence of neuronal dropout in cerebellum were present in homozygotes but not control Townes. These observations suggest that cognitive and behavioral deficits in SCD mice mirror those described in SCD patients and that aging, anemia, and profound neuropathologic changes in hippocampus and cerebellum are possible biologic correlates of those deficits. These findings support using SCD mice for studies of cognitive deficits in SCD and point to vulnerable brain areas with susceptibility to neuronal injury in SCD and to mechanisms that potentially underlie those deficits.


Subject(s)
Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/physiopathology , Cerebellum/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Hippocampus/pathology , Aging/pathology , Aging/physiology , Aging/psychology , Anemia, Sickle Cell/psychology , Animals , Cerebellum/physiopathology , Cognition Disorders/etiology , Cohort Studies , Cross-Sectional Studies , Depression/etiology , Depression/pathology , Depression/physiopathology , Female , Genotype , Hippocampus/physiopathology , Iron/metabolism , Male , Maze Learning/physiology , Mice, Transgenic , Motor Activity/physiology , Neurons/metabolism , Neurons/pathology , Reversal Learning/physiology , Sex Characteristics
7.
Blood Cells Mol Dis ; 55(4): 363-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26460261

ABSTRACT

Fetal hemoglobin-inducing therapies are disease-modifying and ameliorate the pain phenotype in sickle cell disease (SCD). Rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, increases HbF in erythroid precursor cells in vitro. We hypothesized that rapamycin would increase HbF levels and improve nociception phenotype in SCD mice. We used sine-wave electrical stimulation to examine nocifensive phenotype and evaluate myelinated [2000Hz (Aß-fiber) and 250Hz (Aδ-fiber)] and unmyelinated (5Hz C-fibers)] sensory fiber function. Rapamycin significantly increased γ-globin mRNA and HbF levels [+2.3% (0.7, 3.9), mean increase (95% confidence interval, CI), p=0.006]. In homozygous (sickling) mice, long- (16 weeks), but not short-term (6 weeks), rapamycin treatment increased 2000Hz and 250Hz current thresholds in a pattern that varied according to sex. In male, but not female mice, rapamycin (compared with vehicle) was associated with increases in 2000Hz [21Units (7, 35), mean difference (95% CI), p=0.009 for sex∗treatment interaction] and 250Hz [9Units (1, 16), p=0.01] current thresholds. In rapamycin-treated homozygotes, HbF levels directly correlated with myelinated [2000Hz(Aß-fiber, r=0.58, p=0.01) and 250Hz(Aδ-fiber, r=0.6, p=0.01)] but not unmyelinated sensory fiber current thresholds. These findings suggest that in SCD mice, rapamycin increases HbF and modulates current thresholds of myelinated fibers. Therefore, mTOR signaling might be implicated in the pathobiology of SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/physiopathology , Fetal Hemoglobin/biosynthesis , Nociception/drug effects , Phenotype , Sirolimus/pharmacology , Animals , Bone Marrow/pathology , Disease Models, Animal , Female , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Genotype , Male , Mice , Mice, Transgenic , Sensory Thresholds/drug effects , Thermosensing/drug effects
8.
Psychopharmacology (Berl) ; 232(23): 4303-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26337613

ABSTRACT

RATIONALE: Accumulating evidence implicates the nicotinic cholinergic system in autism spectrum disorder (ASD) pathobiology. Neuropathologic studies suggest that nicotinic acetylcholine (ACh) receptor (nAChR) subtypes are altered in brain of autistic individuals. In addition, strategies that increase ACh, the neurotransmitter for nicotinic and muscarinic receptors, appear to improve cognitive deficits in neuropsychiatric disorders and ASD. OBJECTIVE: The aim of this study is to examine the role of the nicotinic cholinergic system on social and repetitive behavior abnormalities and exploratory physical activity in a well-studied model of autism, the BTBR T(+) Itpr3 (tf) /J (BTBR) mouse. METHODS: Using a protocol known to up-regulate expression of brain nAChR subtypes, we measured behavior outcomes before and after BTBR and C57BL/6J (B6) mice were treated (4 weeks) with vehicle or nicotine (50, 100, 200, or 400 µg/ml). RESULTS: Increasing nicotine doses were associated with decreases in water intake, increases in plasma cotinine levels, and at the higher dose (400 µg/ml) with weight loss in BTBR mice. At lower (50, 100 µg/ml) but not higher (200, 400 µg/ml) doses, nicotine increased social interactions in BTBR and B6 mice and at higher, but not lower doses, it decreased repetitive behavior in BTBR. In the open-field test, nicotine at 200 and 400 µg/ml, but not 100 µg/ml compared with vehicle, decreased overall physical activity in BTBR mice. CONCLUSIONS: These findings support the hypotheses that the nicotinic cholinergic system modulates social and repetitive behaviors and may be a therapeutic target to treat behavior deficits in ASD. Further, the BTBR mouse may be valuable for investigations of the role of nAChRs in social deficits and repetitive behavior.


Subject(s)
Autistic Disorder/blood , Cholinergic Agents/pharmacology , Cholinergic Neurons/metabolism , Disease Models, Animal , Interpersonal Relations , Nicotine/pharmacology , Animals , Autistic Disorder/psychology , Cholinergic Neurons/drug effects , Cotinine/blood , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology
9.
Exp Biol Med (Maywood) ; 240(1): 87-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25070860

ABSTRACT

The pain phenotype in sickle cell disease (SCD) patients is highly variable. A small percentage of SCD patients experience many vaso-occlusive crises/year, 5% of patients account for over 30% of pain episodes, while 39% report few episodes of severe pain. Clearly, a better understanding of the pathobiology of SCD is needed to improve its therapy. Humanized sickle cell mice recapitulate several phenotypes of SCD patients and provide a model for the study of SCD pain. Researchers have shown that one strain of humanized SCD mice, the BERK strain, has abnormal pain phenotype. However, the nociception phenotype of another humanized SCD mouse strain, the Townes strain, has not been described. In a large cross-sectional study of BERK and Townes SCD mice, we examined thermosensory response and sensory nerve fiber function using sine-wave electrical stimulation at 2000, 250, and 5 Hz to stimulate preferentially Aß, Aδ, and C sensory nerve fibers, respectively. We found that BERK and Townes mice, compared to respective controls, had decreases in 2000, 250, and 5 Hz current vocalization thresholds in patterns that suggest sensitization of a broad spectrum of sensory nerve fibers. In addition, the pattern of sensitization of sensory fibers varied according to strain, sex, age, and mouse genotype. In a similarly variable pattern, Townes and BERKs also had significantly altered sensitivity to noxious thermal stimuli in agreement with what has been shown by others. In summary, the analysis of somatosensory function using sine-wave electrical stimulation in humanized sickle cell mice suggests that in SCD, both myelinated and unmyelinated, fibers are sensitized. The pattern of sensory fiber sensitization is distinct from that observed in pain models of neuropathic and inflammatory pain. These findings raise the possibility that sensitization of a broad spectrum of sensory fibers might contribute to the altered and variable nociception phenotype in SCD.


Subject(s)
Anemia, Sickle Cell/complications , Nerve Fibers/physiology , Sensation Disorders/etiology , Sensation Disorders/physiopathology , Animals , Cross-Sectional Studies , Disease Models, Animal , Electrophysiological Phenomena , Female , Humans , Male , Mice , Mice, SCID , Mice, Transgenic , Pain/etiology , Pain/physiopathology
10.
Nitric Oxide ; 45: 54-64, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25445633

ABSTRACT

The bioactivity of nitric oxide (NO) is influenced by chemical species generated through reactions with proteins, lipids, metals, and its conversion to nitrite and nitrate. A better understanding of the functions played by each of these species could be achieved by developing selective assays able of distinguishing nitrite from other NO species. Nagababu and Rifkind developed a method using acetic and ascorbic acids to measure nitrite-derived NO in plasma. Here, we adapted, optimized, and validated this method to assay nitrite in tissues. The method yielded linear measurements over 1-300 pmol of nitrite and was validated for tissue preserved in a nitrite stabilization solution composed of potassium ferricyanide, N-ethylmaleimide and NP-40. When samples were processed with chloroform, but not with methanol, ethanol, acetic acid or acetonitrile, reliable and reproducible nitrite measurements in up to 20 sample replicates were obtained. The method's accuracy in tissue was ≈ 90% and in plasma 99.9%. In mice, during basal conditions, brain, heart, lung, liver, spleen and kidney cortex had similar nitrite levels. In addition, nitrite tissue levels were similar regardless of when organs were processed: immediately upon collection, kept in stabilization solution for later analysis or frozen and later processed. After ip nitrite injections, rapidly changing nitrite concentrations in tissue and plasma could be measured and were shown to change in significantly distinct patterns. This validated method could be valuable for investigations of nitrite biology in conditions such as sickle cell disease, cardiovascular disease, and diabetes, where nitrite is thought to play a role.


Subject(s)
Clinical Chemistry Tests/methods , Histocytochemistry/methods , Nitrites/analysis , Acetic Acid/chemistry , Animals , Ascorbic Acid/chemistry , Clinical Chemistry Tests/standards , Female , Histocytochemistry/standards , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitrites/metabolism , Organ Specificity , Reproducibility of Results , Sensitivity and Specificity
11.
PLoS One ; 7(11): e48733, 2012.
Article in English | MEDLINE | ID: mdl-23139814

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is a fatal neurodegenerative disorder caused by a deficiency of palmitoyl-protein thioesterase-1 (PPT1). We have previously shown that children with INCL have increased risk of hypothermia during anesthesia and that PPT1-deficiency in mice is associated with disruption of adaptive energy metabolism, downregulation of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), and mitochondrial dysfunction. Here we hypothesized that Ppt1-knockout mice, a well-studied model of INCL that shows many of the neurologic manifestations of the disease, would recapitulate the thermoregulation impairment observed in children with INCL. We also hypothesized that when exposed to cold, Ppt1-knockout mice would be unable to maintain body temperature as in mice thermogenesis requires upregulation of Pgc-1α and uncoupling protein 1 (Ucp-1) in brown adipose tissue. We found that the Ppt1-KO mice had lower basal body temperature as they aged and developed hypothermia during cold exposure. Surprisingly, this inability to maintain body temperature during cold exposure in Ppt1-KO mice was associated with an adequate upregulation of Pgc-1α and Ucp-1 but with lower levels of sympathetic neurotransmitters in brown adipose tissue. In addition, during baseline conditions, brown adipose tissue of Ppt1-KO mice had less vacuolization (lipid droplets) compared to wild-type animals. After cold stress, wild-type animals had significant decreases whereas Ppt1-KO had insignificant changes in lipid droplets compared with baseline measurements, thus suggesting that Ppt1-KO had less lipolysis in response to cold stress. These results uncover a previously unknown phenotype associated with PPT1 deficiency, that of altered thermoregulation, which is associated with impaired lipolysis and neurotransmitter release to brown adipose tissue during cold exposure. These findings suggest that INCL should be added to the list of neurodegenerative diseases that are linked to alterations in peripheral metabolic processes. In addition, extrapolating these findings clinically, impaired thermoregulation and hypothermia are potential risks in patients with INCL.


Subject(s)
Adipose Tissue, Brown/abnormalities , Adipose Tissue, Brown/physiopathology , Body Temperature Regulation/physiology , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Thiolester Hydrolases/deficiency , Adenosine Triphosphate/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Cold Temperature , Disease Models, Animal , Female , Hot Temperature , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , NAD/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neurotransmitter Agents/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Adrenergic, beta/metabolism , Thiolester Hydrolases/metabolism , Uncoupling Protein 1 , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology , Ventromedial Hypothalamic Nucleus/physiopathology
12.
Nitric Oxide ; 26(2): 81-8, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22202903

ABSTRACT

Nitric oxide synthases (NOSs) have been shown to modulate thermal hyperalgesia and mechanical hypersensitivity in inflammatory and neuropathic pain. However, little is known about the effect of NOSs on baseline function of sensory nerve fibers. Using genetic deficiency and pharmacologic inhibition of NOSs, we examined the impact of the three isoforms NOS1, NOS2, and NOS3 on baseline nocifensive behavior by measuring current vocalization threshold in response to electrical stimulation at 5, 250, 2000 Hz that preferentially stimulate C, Aδ, and Aß fibers. In response to 5, 250 and 2000 Hz, NOS1-deficient animals had significantly higher current vocalization thresholds compared with wild-type. Genetic deficiency of NOS2 was associated with higher current vocalization thresholds in response to 5 Hz (C-fiber) stimulation. In contrast, NOS3-deficient animals had an overall weak trend toward lower current vocalization thresholds at 5 Hz and significantly lower current vocalization threshold compared with wild-type animals at 250 and 2000 Hz. Therefore, NOSs distinctively affect baseline mouse current vocalization threshold and appear to play a role on nocifensive response to electrical stimulation of sensory nerve fibers.


Subject(s)
Nitric Oxide Synthase/metabolism , Nociception/physiology , Vocalization, Animal/physiology , Analysis of Variance , Animals , Brain Chemistry , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , Isoenzymes , Male , Mice , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/analysis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitrites/analysis , Pain Measurement , Spinal Cord/chemistry , Spinal Cord/metabolism
13.
Anesthesiology ; 114(5): 1190-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21383614

ABSTRACT

BACKGROUND: Previous studies suggest that the transient receptor potential vanilloid 1 (TRPV1) channel has a role in sepsis, but it is unclear whether its effect on survival and immune response is beneficial or harmful. METHODS: We studied the effects of genetic (Trpv1-knockout vs. wild-type [WT] mice) and pharmacologic disruption of TRPV1 with resiniferatoxin (an agonist) or capsazepine (an antagonist) on mortality, bacterial clearance, and cytokine expression during lipopolysaccharide or cecal ligation and puncture-induced sepsis. RESULTS: After cecal ligation and puncture, genetic disruption of TRPV1 in Trpv1-knockout versus WT mice was associated with increased mortality risk (hazard ratio, 2.17; 95% CI, 1.23-3.81; P = 0.01). Furthermore, pharmacologic disruption of TRPV1 with intrathecal resiniferatoxin, compared with vehicle, increased mortality risk (hazard ratio, 1.80; 95% CI, 1.05-3.2; P = 0.03) in WT, but not in Trpv1-knockout, mice. After lipopolysaccharide, neither genetic (Trpv1 knockout) nor pharmacologic disruption of TRPV1 with resiniferatoxin had significant effect on survival compared with respective controls. In contrast, after lipopolysaccharide, pharmacologic disruption of TRPV1 with capsazepine, compared with vehicle, increased mortality risk (hazard ratio, 1.92; 95% CI, 1.02-3.61; P = 0.04) in WT animals. Furthermore, after cecal ligation and puncture, increased mortality in resiniferatoxin-treated WT animals was associated with higher blood bacterial count (P = 0.0004) and higher nitrate/nitrite concentrations and down-regulation of tumor necrosis factor α expression (P = 0.004) compared with controls. CONCLUSIONS: Genetic or pharmacologic disruption of TRPV1 can affect mortality, blood bacteria clearance, and cytokine response in sepsis in patterns that may vary according to the sepsis-inducing event and the method of TRPV1 disruption.


Subject(s)
Bacterial Load/drug effects , Cytokines/drug effects , Gene Expression/genetics , Sepsis/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Animals , Bacterial Load/genetics , Capsaicin/administration & dosage , Capsaicin/analogs & derivatives , Cecum/surgery , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Diterpenes/administration & dosage , Down-Regulation , Female , Flow Cytometry , Gene Expression/drug effects , Ligation , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Lavage , Peritoneum/drug effects , Peritoneum/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/genetics , Survival Analysis , TRPV Cation Channels/drug effects , Tumor Necrosis Factor-alpha
14.
J Neurosci Res ; 88(2): 248-57, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19705456

ABSTRACT

Vascular endothelial growth factor (VEGF) is an endothelial and neuronal survival factor and a mitogen for endothelial cells and astrocytes in both explant and in vivo injury models. In the CNS, interplay between the vasculature and neural stem progenitor (NSP) cells is required for the maintenance of angiogenic/neurogenic coordination in the germinal niche in the subventricular zone (SVZ) of the lateral ventricle. Using an in vitro SVZ neurosphere (NS) model, this study aimed to understand the direct effects of VEGF and its receptor signaling on neonatal NSP cell growth and migration. Our data indicate that VEGF administration, compared with untreated or brain-derived neurotrophic factor-treated NS, significantly increased growth and migratory capacity of glial fibrillary acidic protein (GFAP)(+) and nestin(+) NSP cells and in secondary cultures induced a stellate astrocyte morphology. Blockade of both VEGF, which is normally expressed in some NS cells, and its flt-1 receptor signaling by neutralizing antibodies caused morphological changes specifically in GFAP(+) cells and disrupted sphere formation and outward migration. These cells did not appear as conventional polygonal astrocytes; their process growth was severely restricted, and overall migration was reduced by up to 76% of control cultures. Blockade of VEGF's flk-1 receptor reduced VEGF expression and caused a lesser, though significant, decrease (29%) in NSP (GFAP(+)) cell migration. The results show that both VEGF and, in particular, flt-1 receptor signaling are critical to the proper configuration of the NS and its subsequent development. VEGF is also an important growth and migratory factor particularly for GFAP(+) cells developing in SVZ-derived NS in culture.


Subject(s)
Astrocytes/physiology , Cell Movement/physiology , Stem Cell Niche/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Astrocytes/cytology , Brain-Derived Neurotrophic Factor/metabolism , Cell Enlargement , Cells, Cultured , Cerebral Ventricles , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
J Nucl Med ; 49(2): 295-302, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199622

ABSTRACT

UNLABELLED: The aim of this study was to determine if pulsed high-intensity focused ultrasound (HIFU) exposures could enhance tumor uptake of (111)In-MX-B3, a murine IgG1kappa monoclonal antibody directed against the Le(y) antigen. METHODS: MX-B3 was labeled with (111)In, purified, and confirmed for its binding to the antigen-positive A431 cell line. Groups of nude mice were inoculated subcutaneously with A431 tumor cells on both hind flanks. A tumor on one flank was treated with pulsed-HIFU; the other tumor was used as an untreated control. Within 10 min after the HIFU exposure, the mice received intravenous (111)In-MX-B3 for imaging and biodistribution studies. Mice were euthanized at 1, 24, 48, and 120 h after injection for biodistribution studies. RESULTS: The HIFU exposure shortened the peak tumor uptake time (24 vs. 48 h for the control) and increased the peak tumor uptake value (38 vs. 25 %ID/g [percentage injected dose per gram] for the control). The HIFU effect on enhancing tumor uptake was greater at earlier times up to 24 h, but the effect was gradually diminished thereafter. The HIFU effect on enhancing tumor uptake was substantiated by nuclear imaging studies. HIFU also increased the uptake of the antibody in surrounding tissues, but the net increase was marginal compared with the increase in tumor uptake. CONCLUSION: This study demonstrates that pulsed-HIFU significantly enhances the delivery of (111)In-MX-B3 in human epidermoid tumors xenografted in nude mice. The results of this pilot study warrant further evaluation of other treatment regimens, such as repeated HIFU exposures for greater delivery enhancement of antibodies labeled with cytotoxic radioisotopes or pulsed-HIFU exposure in addition to a combined therapy of (90)Y-B3 and taxol to enhance the synergistic effect.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Phonophoresis/methods , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/therapy , Humans , Isotope Labeling , Metabolic Clearance Rate/radiation effects , Mice , Mice, Nude , Organ Specificity/radiation effects , Radioimmunotherapy/methods , Radionuclide Imaging , Tissue Distribution/radiation effects
16.
Intensive Care Med ; 33(11): 1993-2003, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17684724

ABSTRACT

OBJECTIVE: To investigate the role of neuronal nitric oxide synthase (NOS1) in murine polymicrobial peritonitis and sepsis. DESIGN: Randomized experimental trial. SETTING: Animal research facility. SUBJECTS: B6129S NOS1+/+ and B6;129S4 NOS-/- mice. INTERVENTIONS: NOS1+/+ and NOS1-/- animals underwent cecal ligation and puncture (CLP) or sham surgery and received the NOS1 inhibitor 7-nitroindazole (7-NI) or vehicle. MEASUREMENTS AND MAIN RESULTS: After CLP, genetic deficiency and pharmacologic inhibition of NOS1 significantly increased risk of mortality [8.69 (3.27, 23.1), p<0.0001 and 1.71 (1.00, 2.92) p=0.05, hazard ratio of death (95% confidence interval) for NOS1-/- and 7-NI-treated NOS1+/+ respectively] compared with NOS1+/+ animals. In 7-NI-treated NOS1+/+ animals, there were increases (6 h) and then decreases (24 h), whereas in NOS-/- animals persistent increases in blood bacteria counts (p=0.04 for differing effects of 7-NI and NOS1-/-) were seen compared with NOS1(+/+) animals. After CLP, NOS1(-/-) had upregulation of inducible NOS and proinflammatory cytokines and greater increases in serum tumor necrosis factor-alpha and interleukin-6 levels compared with NOS1+/+ mice (all p<0.05). Following CLP, there were similar significant decreases in circulating leukocytes and lung lavage cells (p

Subject(s)
Nitric Oxide Synthase Type I/deficiency , Peritonitis/physiopathology , Sepsis/physiopathology , Animals , Female , Gene Expression , Mice , Nitric Oxide Synthase Type I/adverse effects , Nitric Oxide Synthase Type I/analysis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , Peritonitis/mortality , Random Allocation , Risk Assessment , Sepsis/mortality , Survival Analysis , United States
17.
Exp Neurol ; 192(2): 394-406, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15755557

ABSTRACT

The non-angiogenic role of vascular endothelial growth factor (VEGF), and its receptors flt-1 and flk-1, together with downstream signaling pathways were examined in fetal and postnatal rat cerebral cortical organotypic explants. VEGF application in both paradigms caused a significant increase in astroglial proliferation and a dose-dependent increase in GFAP and nestin immunoreactivity. The VEGF receptor flt-1 was observed on most, though not all astrocytes, while flk-1 receptor immunoexpression was absent. Treatment with antisense oligonucleotides (AS-ODNs) to flt-1 resulted in a dramatic decrease in GFAP and nestin immunoreactivity, which further confirmed the role of flt-1 in mediating VEGF's gliotrophic effects, while AS-ODNs to flk-1 had no effect. VEGF-induced gliotrophic effects were found to be mediated by the MAPK/ERK and PI-3 kinase signaling pathways, since the both the MEK1 inhibitor, PD98059 and the PI-3 kinase inhibitor, Wortmannin abolished VEGF-induced astrocytic GFAP(+) expression. Although high dose VEGF application resulted in strong upregulation of both GFAP and nestin immunoreactivity in astrocytes, overlap of the two proteins was not observed in all cells, suggesting that some of the nestin(+) cells might be neural progenitors. Exposure to VEGF resulted in upregulation of both VEGF and bFGF mRNA at the one-day time point, and bFGF protein by 3 days; VEGF activated astrocytes expressed bFGF to a much greater degree than those in untreated explants. The increased expression of bFGF induced by VEGF, may serve in the proliferation of multipotential neural stem/progenitor cells in vitro. VEGF, an established angiogenic factor, appears to play a significant role in the growth and differentiation of astrocytes in the CNS.


Subject(s)
Astrocytes/drug effects , Neocortex/cytology , Proteins/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Blotting, Northern/methods , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Intermediate Filament Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neocortex/drug effects , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Nestin , Oligodeoxyribonucleotides, Antisense/pharmacology , Organ Culture Techniques/methods , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Brain Res Dev Brain Res ; 148(1): 59-68, 2004 Jan 31.
Article in English | MEDLINE | ID: mdl-14757519

ABSTRACT

Recent studies have demonstrated that vascular endothelial growth factor (VEGF) and its receptor VEGFR2 (flk-1) are expressed by neurons during development and following hypoxic-ischemic events. Moreover, fetal CNS tissue explants exposed to exogenous VEGF exhibit increased neuronal Map-2 expression, suggesting that VEGF could have an effect on neuronal maturation. To determine whether this effect is of a direct nature, we examined the expression of Map-2 in the presence of VEGF in primary CNS neuronal cultures. After 3 days in culture, a statistically significant dose-dependent increase in the length of Map-2(+) processes was observed, with the peak occurring at 10 ng/ml of VEGF. Immunohistochemical analysis of the cultures demonstrated the presence of VEGFR2 after VEGF treatment, as well as the expression of the VEGF receptor VEGFR1 (flt-1). Treatment of the cultures with antisense oligonucleotides against VEGFR2, but not against VEGFR1, abolished the effect of VEGF on the length of Map-2(+) processes. RT-PCR analyses of Map-2 and VEGFR1 indicated that mRNAs of these two genes are upregulated in the presence of VEGF. The addition of wortmannin, an inhibitor of PI3K/Akt signal-transduction pathway, to the media did not affect the VEGF-dependent increase in Map-2(+) length. In contrast PD98059, which inhibits the MAPK pathway, partially abolished this effect of VEGF. These experiments suggest that VEGF has a direct effect on neuronal growth and maturation under normoxic conditions during CNS development, which is mediated by the VEGFR2 receptor via the MAPK pathway.


Subject(s)
Central Nervous System/cytology , Neurites/drug effects , Neurons/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Androstadienes/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Immunohistochemistry/methods , Microtubule-Associated Proteins/metabolism , Neurites/physiology , Neurons/physiology , Oligonucleotides, Antisense/pharmacology , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Tubulin/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wortmannin
19.
J Cereb Blood Flow Metab ; 23(12): 1420-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14663337

ABSTRACT

SUMMARY: The angiogenic role of vascular endothelial growth factor (VEGF) receptors, flk-1 and flt-1, and their downstream signaling pathways, MAPK/ERK and PI-3 kinase, were examined in a fetal rat cortical explant model after exposure to exogenous VEGF. Treatment with VEGF resulted in substantial neovascularization characterized by increased vascular flk-1 receptor expression, whereas flt-1 receptor protein expression was absent. The specific role of flk-1 receptors in the angiogenic process was confirmed by the addition of antisense oligonucleotides (AS-ODNs) to flk-1, which blocked angiogenesis, whereas AS-ODNs to flt-1 had no effect. These results were further supported by the finding that specific chemical inhibition of flk-1 receptors caused disruption of the angiogenic response, whereas inhibition of the flt-1 receptors had no effect. Application of either MAPK/ERK or PI-3 kinase pathway inhibitors disrupted VEGF-induced angiogenesis, thereby indicating that both signaling pathways mediate this process. Thus VEGF binding to the endothelial flk-1 receptor activates the MAPK/ERK and PI-3 kinase pathways, resulting in neoangiogenic events. Of interest is the fact that although VEGF is regarded as a vascular permeability factor, its application to nascent cortical tissue caused an increase in a key physiologic protein of the blood-brain barrier function, glucose transporter-1, suggesting that the cytokine may have a role in blood-brain barrier development.


Subject(s)
Brain Tissue Transplantation , Fetal Tissue Transplantation , MAP Kinase Signaling System/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Blood-Brain Barrier/physiology , Cells, Cultured , Endothelium, Vascular/metabolism , Female , Glucose Transporter Type 1 , MAP Kinase Signaling System/drug effects , Monosaccharide Transport Proteins/metabolism , Neovascularization, Physiologic/drug effects , Neurons/cytology , Neurons/transplantation , Pregnancy , Rats , Rats, Wistar , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
J Neurosci ; 23(35): 11036-44, 2003 Dec 03.
Article in English | MEDLINE | ID: mdl-14657160

ABSTRACT

Vascular endothelial growth factor (VEGF) is well known to play an important regulatory role in vascular growth and development. Because gene knock-outs of VEGF and its receptors flk-1 and flt-1 result in early embryonic lethality, determining roles for VEGF in CNS development has been particularly difficult. Recent studies have shown that VEGF is upregulated after various injuries to the adult brain and that the cytokine affords protection to cultured neurons affected by oxidative or excitotoxic stress. The present study demonstrates, for the first time, that VEGF is directly neurotrophic to CNS neurons in culture. We applied VEGF to normoxic fetal organotypic cortical explants as a model of CNS neuropil, in addition to primary cortical neurons, to assess direct growth effects absent vascular or astroglial activity. We found that VEGF provided a significant dose-responsive increase in the neuronal microtubule markers TUJ1 and MAP-2, as well as mRNA for MAP-2 and flk-1. Antisense oligodeoxynucleotides to flk-1, but not flt-1, inhibited neuritic outgrowth, whereas inhibitors of the signaling pathways MEK1 and P13-AKT both abrogated VEGF-induced growth. VEGF applied to primary cortical neurons produced significant increases in neuronal cell body diameter and the number of emerging neurites mediated by flk-1. Possibly, VEGF achieves its effects by acting on the neuronal microtubular content, which is involved with growth, stability and maturation. Several studies have now shown that VEGF is neurotrophic and neuroprotective independent of a vascular component; we suggest that VEGF plays seminal pleiotrophic roles in CNS development and repair.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/drug effects , Protein Serine-Threonine Kinases , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Culture Media, Serum-Free/pharmacology , Gene Expression Regulation, Developmental/drug effects , In Vitro Techniques , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Signal Transduction/drug effects , Tubulin/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...