Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Ecancermedicalscience ; 17: 1582, 2023.
Article in English | MEDLINE | ID: mdl-37533941

ABSTRACT

99mTc-EDDA/HYNIC-TOC is an easily available and cheaper radionuclide that could be used for somatostatin-receptor-based imaging of neuroendocrine tumours (NETs). We aimed to evaluate the diagnostic performance of 99mTc-EDDA/HYNIC-TOC compared to111In-DTPA-octreotide in patients (pts) with NETs. We performed a prospective diagnostic study including pts with biopsy-confirmed NET and at least one visible lesion at conventional imaging. Two independent nuclear medicine physicians evaluated pts who underwent 99mTc and 111In scans and images. The primary outcome was comparative diagnostic accuracy of 99mTc and 111In. Secondary outcomes include safety. Nine pts were included and performed 14 paired scans. Overall, 126 lesions were identified. 99mTc demonstrated superior sensitivity both when all images were analysed (93.7, 95% CI 88.1% - 96.8% versus 74.8%, 95% CI 66.6 - 81.6%, p < 0.001) and when liver-specific images were analysed (97.8%, 95% CI 92.7% - 99.5% versus 85.1%, 95% CI 76.6% - 91.0%, p < 0.001). 99mTc was also associated with a lower negative likelihood ratio (LR) (0.002, 95% CI 0.009 - 0.1 versus 0.19, 95% CI 0.12 - 0.42, p = 0.009) when evaluating hepatic lesions. Adverse events happened in 3 pts after 111In and in 2 pts after 99mTc, all grade 1. The 99mTc demonstrated a higher sensitivity overall and a better negative LR in liver-specific images compared to 111In in pts with NETs. Our findings suggest that 99mTc is an alternative to 111In and is especially useful in ruling out liver metastases. NCT02691078.

2.
Brain Res Bull ; 200: 110694, 2023 08.
Article in English | MEDLINE | ID: mdl-37353036

ABSTRACT

AIMS: We have previously demonstrated that fructose supplementation (FS), given in a scheme used for inducing metabolic syndrome (MS), elicited pain relief in the nitroglycerin (NTG)-elicited rat migraine model. Herein, we evaluated whether FS could reestablish the impaired metabolic pathways in NTG-injected rats. MAIN METHODS: Male Wistar rats (N = 40) were divided into two groups for receiving 10 % FS or tap water. After 45 days, they were subdivided into NTG-injected (10 mg/kg; 15 days) or controls. After the fourth NTG injection, 18F-fluorodeoxyglucose ([18F] FDG) micro-PET scanning was accomplished. The day after, euthanasia was performed, and blood was collected for glycemia and LDH analysis. The levels of energy molecules, TBARS, PGC-1α, and MCTS1 were evaluated in the brain cortices. The activated satellite glial cells (SGC) were assessed in the trigeminal ganglion (TG). KEY FINDINGS: There were no variations of glycemia or LDH serum levels. NTG-injected rats showed a significant increase in glucose uptake in the hypothalamus (HT) vs. NTG-free rats. The FS-NTG group showed increased metabolism in the superior colliculus (SC) vs. the NTG group. Moreover, the glucose uptake was amplified in the inferior colliculus (IC) of the FS-NTG vs. FS group. The cortical inosine levels were significantly higher in FS-NTG rats vs. NTG or FS groups, with no changes in TBARS or MCTS1 levels, despite a minor decrease of PGC1-α contents in the FS+NTG group. Finally, there was a significant increase of activated SGC around TG in the FS-NTG rats. SIGNIFICANCE: We provide novel evidence linking nutrition and metabolism with migraine.


Subject(s)
Fructose , Migraine Disorders , Rats , Male , Animals , Rats, Wistar , Fructose/pharmacology , Thiobarbituric Acid Reactive Substances , Migraine Disorders/chemically induced , Nitroglycerin/pharmacology , Brain/metabolism , Dietary Supplements , Glucose , Disease Models, Animal
3.
Eur J Nucl Med Mol Imaging ; 49(7): 2251-2264, 2022 06.
Article in English | MEDLINE | ID: mdl-35122511

ABSTRACT

PURPOSE: Advances in functional imaging allowed us to visualize brain glucose metabolism in vivo and non-invasively with [18F]fluoro-2-deoxyglucose (FDG) positron emission tomography (PET) imaging. In the past decades, FDG-PET has been instrumental in the understanding of brain function in health and disease. The source of the FDG-PET signal has been attributed to neuronal uptake, with hypometabolism being considered as a direct index of neuronal dysfunction or death. However, other brain cells are also metabolically active, including astrocytes. Based on the astrocyte-neuron lactate shuttle hypothesis, the activation of the glutamate transporter 1 (GLT-1) acts as a trigger for glucose uptake by astrocytes. With this in mind, we investigated glucose utilization changes after pharmacologically downregulating GLT-1 with clozapine (CLO), an anti-psychotic drug. METHODS: Adult male Wistar rats (control, n = 14; CLO, n = 12) received CLO (25/35 mg kg-1) for 6 weeks. CLO effects were evaluated in vivo with FDG-PET and cortical tissue was used to evaluate glutamate uptake and GLT-1 and GLAST levels. CLO treatment effects were also assessed in cortical astrocyte cultures (glucose and glutamate uptake, GLT-1 and GLAST levels) and in cortical neuronal cultures (glucose uptake). RESULTS: CLO markedly reduced in vivo brain glucose metabolism in several brain areas, especially in the cortex. Ex vivo analyses demonstrated decreased cortical glutamate transport along with GLT-1 mRNA and protein downregulation. In astrocyte cultures, CLO decreased GLT-1 density as well as glutamate and glucose uptake. By contrast, in cortical neuronal cultures, CLO did not affect glucose uptake. CONCLUSION: This work provides in vivo demonstration that GLT-1 downregulation induces astrocyte-dependent cortical FDG-PET hypometabolism-mimicking the hypometabolic signature seen in people developing dementia-and adds further evidence that astrocytes are key contributors of the FDG-PET signal.


Subject(s)
Astrocytes , Clozapine , Animals , Clozapine/metabolism , Clozapine/pharmacology , Fluorodeoxyglucose F18/metabolism , Glucose/metabolism , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Male , Positron-Emission Tomography , Rats , Rats, Wistar
4.
Metab Brain Dis ; 37(4): 911-926, 2022 04.
Article in English | MEDLINE | ID: mdl-35059965

ABSTRACT

Allergic asthma is characterized by chronic airway inflammation and is constantly associated with anxiety disorder. Recent studies showed bidirectional interaction between the brain and the lung tissue. However, where and how the brain is affected in allergic asthma remains unclear. We aimed to investigate the neuroinflammatory, neurochemical, and neurometabolic alterations that lead to anxiety-like behavior in an experimental model of allergic asthma. Mice were submitted to an allergic asthma model induced by ovalbumin (OVA) and the control group received only Dulbecco's phosphate-buffered saline (DPBS). Our findings indicate that airway inflammation increases interleukin (IL) -9, IL-13, eotaxin, and IL-1ß release and changes acetylcholinesterase (AChE) and Na+,K+-ATPase activities in the brain of mice. Furthermore, we demonstrate that a higher reactive oxygen species (ROS) formation and antioxidant defense alteration that leads to protein damage and mitochondrial dysfunction. Therefore, airway inflammation promotes a pro-inflammatory environment with an increase of BDNF expression in the brain of allergic asthma mice. These pro-inflammatory environments lead to an increase in glucose uptake in the limbic regions and to anxiety-like behavior that was observed through the elevated plus maze (EPM) test and downregulation of glucocorticoid receptor (GR). In conclusion, the present study revealed for the first time that airway inflammation induces neuroinflammatory, neurochemical, and neurometabolic changes within the brain that leads to anxiety-like behavior. Knowledge about mechanisms that lead to anxiety phenotype in asthma is a beneficial tool that can be used for the complete management and treatment of the disease.


Subject(s)
Acetylcholinesterase , Asthma , Animals , Anxiety , Asthma/chemically induced , Disease Models, Animal , Inflammation/chemically induced , Inflammation/metabolism , Mice
5.
Neurotox Res ; 39(6): 1830-1845, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34797528

ABSTRACT

Methylphenidate (MPH) has been widely misused by children and adolescents who do not meet all diagnostic criteria for attention-deficit/hyperactivity disorder without a consensus about the consequences. Here, we evaluate the effect of MPH treatment on glucose metabolism and metabolic network in the rat brain, as well as on performance in behavioral tests. Wistar male rats received intraperitoneal injections of MPH (2.0 mg/kg) or an equivalent volume of 0.9% saline solution (controls), once a day, from the 15th to the 44th postnatal day. Fluorodeoxyglucose-18 was used to investigate cerebral metabolism, and a cross-correlation matrix was used to examine the brain metabolic network in MPH-treated rats using micro-positron emission tomography imaging. Performance in the light-dark transition box, eating-related depression, and sucrose preference tests was also evaluated. While MPH provoked glucose hypermetabolism in the auditory, parietal, retrosplenial, somatosensory, and visual cortices, hypometabolism was identified in the left orbitofrontal cortex. MPH-treated rats show a brain metabolic network more efficient and connected, but careful analyses reveal that the MPH interrupts the communication of the orbitofrontal cortex with other brain areas. Anxiety-like behavior was also observed in MPH-treated rats. This study shows that glucose metabolism evaluated by micro-positron emission tomography in the brain can be affected by MPH in different ways according to the region of the brain studied. It may be related, at least in part, to a rewiring in the brain the metabolic network and behavioral changes observed, representing an important step in exploring the mechanisms and consequences of MPH treatment.


Subject(s)
Anxiety/chemically induced , Glucose/metabolism , Methylphenidate/pharmacology , Prefrontal Cortex/drug effects , Animals , Anxiety/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Homeostasis/drug effects , Male , Metabolic Networks and Pathways/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
6.
Front Psychiatry ; 12: 701408, 2021.
Article in English | MEDLINE | ID: mdl-34421682

ABSTRACT

Major depressive disorder (MDD) leads to pervasive changes in the health of afflicted patients. Despite advances in the understanding of MDD and its treatment, profound innovation is needed to develop fast-onset antidepressants with higher effectiveness. When acutely administered, the endogenous nucleoside guanosine (GUO) shows fast-onset antidepressant-like effects in several mouse models, including the olfactory bulbectomy (OBX) rodent model. OBX is advocated to possess translational value and be suitable to assess the time course of depressive-like behavior in rodents. This study aimed at investigating the long-term behavioral and neurochemical effects of GUO in a mouse model of depression induced by bilateral bulbectomy (OBX). Mice were submitted to OBX and, after 14 days of recovery, received daily (ip) administration of 7.5 mg/kg GUO or 40 mg/kg imipramine (IMI) for 45 days. GUO and IMI reversed the OBX-induced hyperlocomotion and recognition memory impairment, hippocampal BDNF increase, and redox imbalance (ROS, NO, and GSH levels). GUO also mitigated the OBX-induced hippocampal neuroinflammation (IL-1, IL-6, TNF-α, INF-γ, and IL-10). Brain microPET imaging ([18F]FDG) shows that GUO also prevented the OBX-induced increase in hippocampal FDG metabolism. These results provide additional evidence for GUO antidepressant-like effects, associated with beneficial neurochemical outcomes relevant to counteract depression.

7.
Toxicology ; 454: 152737, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33631299

ABSTRACT

Ureases are microbial virulence factors either because of the enzymatic release of ammonia or due to many other non-enzymatic effects. Here we studied two neurotoxic urease isoforms, Canatoxin (CNTX) and Jack Bean Urease (JBU), produced by the plant Canavalia ensiformis, whose mechanisms of action remain elusive. The neurotoxins provoke convulsions in rodents (LD50 ∼2 mg/kg) and stimulate exocytosis in cell models, affecting intracellular calcium levels. Here, electrophysiological and brain imaging techniques were applied to elucidate their mode of action. While systemic administration of the toxins causes tonic-clonic seizures in rodents, JBU injected into rat hippocampus induced spike-wave discharges similar to absence-like seizures. JBU reduced the amplitude of compound action potential from mouse sciatic nerve in a tetrodotoxin-insensitive manner. Hippocampal slices from CNTX-injected animals or slices treated in vitro with JBU failed to induce long term potentiation upon tetanic stimulation. Rat cortical synaptosomes treated with JBU released L-glutamate. JBU increased the intracellular calcium levels and spontaneous firing rate in rat hippocampus neurons. MicroPET scans of CNTX-injected rats revealed increased [18]Fluoro-deoxyglucose uptake in epileptogenesis-related areas like hippocampus and thalamus. Curiously, CNTX did not affect voltage-gated sodium, calcium or potassium channels currents, neither did it interfere on cholinergic receptors, suggesting an indirect mode of action that could be related to the ureases' membrane-disturbing properties. Understanding the neurotoxic mode of action of C. ensiformis ureases could help to unveil the so far underappreciated relevance of these toxins in diseases caused by urease-producing microorganisms, in which the human central nervous system is affected.


Subject(s)
Canavalia/chemistry , Neurotoxicity Syndromes/etiology , Plant Proteins/toxicity , Toxins, Biological/toxicity , Urease/toxicity , Animals , Convulsants/isolation & purification , Convulsants/toxicity , Female , Male , Mice , Nervous System/drug effects , Nervous System/pathology , Neurotoxicity Syndromes/physiopathology , Plant Proteins/isolation & purification , Rats , Rats, Wistar , Toxins, Biological/isolation & purification , Urease/isolation & purification , Xenopus laevis
8.
J Neurochem ; 157(6): 1911-1929, 2021 06.
Article in English | MEDLINE | ID: mdl-33098090

ABSTRACT

Prenatal and early postnatal periods are important for brain development and neural function. Neonatal insults such as hypoxia-ischemia (HI) causes prolonged neural and metabolic dysregulation, affecting central nervous system maturation. There is evidence that brain hypometabolism could increase the risk of adult-onset neurodegenerative diseases. However, the impact of non-pharmacologic strategies to attenuate HI-induced brain glucose dysfunction is still underexplored. This study investigated the long-term effects of early environmental enrichment in metabolic, cell, and functional responses after neonatal HI. Thereby, male Wistar rats were divided according to surgical procedure, sham, and HI (performed at postnatal day 3), and the allocation to standard (SC) or enriched condition (EC) during gestation and lactation periods. In-vivo cerebral metabolism was assessed by means of [18 F]-FDG micro-positron emission tomography, and cognitive, biochemical, and histological analyses were performed in adulthood. Our findings reveal that HI causes a reduction in glucose metabolism and glucose transporter levels as well as hyposynchronicity in metabolic brain networks. However, EC during prenatal or early postnatal period attenuated these metabolic disturbances. A positive correlation was observed between [18 F]-FDG values and volume ratios in adulthood, indicating that preserved tissue by EC is metabolically active. EC promotes better cognitive scores, as well as down-regulation of amyloid precursor protein in the parietal cortex and hippocampus of HI animals. Furthermore, growth-associated protein 43 was up-regulated in the cortex of EC animals. Altogether, results presented support that EC during gestation and lactation period can reduce HI-induced impairments that may contribute to functional decline and progressive late neurodegeneration.


Subject(s)
Brain/metabolism , Environment , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , Neuronal Plasticity/physiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Female , Hypoxia-Ischemia, Brain/psychology , Lactation/metabolism , Lactation/psychology , Male , Maze Learning/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neurodegenerative Diseases/psychology , Positron-Emission Tomography/methods , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Wistar
9.
Am J Physiol Endocrinol Metab ; 319(5): E877-E892, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32893672

ABSTRACT

Free fatty acid (FFA) receptors FFA1 and FFA4 are omega-3 molecular targets in metabolic diseases; however, their function in cancer cachexia remains unraveled. We assessed the role of FFA1 and FFA4 receptors in the mouse model of cachexia induced by Lewis lung carcinoma (LLC) cell implantation. Naturally occurring ligands such as α-linolenic acid (ALA) and docosahexaenoic acid (DHA), the synthetic FFA1/FFA4 agonists GW9508 and TUG891, or the selective FFA1 GW1100 or FFA4 AH7614 antagonists were tested. FFA1 and FFA4 expression and other cachexia-related parameters were evaluated. GW9508 and TUG891 decreased tumor weight in LLC-bearing mice. Regarding cachexia-related end points, ALA, DHA, and the preferential FFA1 agonist GW9508 rescued body weight loss. Skeletal muscle mass was reestablished by ALA treatment, but this was not reflected in the fiber cross-sectional areas (CSA) measurement. Otherwise, TUG891, GW1100, or AH7614 reduced the muscle fiber CSA. Treatments with ALA, GW9508, GW1100, or AH7614 restored white adipose tissue (WAT) depletion. As for inflammatory outcomes, ALA improved anemia, whereas GW9508 reduced splenomegaly. Concerning behavioral impairments, ALA and GW9508 rescued locomotor activity, whereas ALA improved motor coordination. Additionally, DHA improved grip strength. Notably, GW9508 restored abnormal brain glucose metabolism in different brain regions. The GW9508 treatment increased leptin levels, without altering uncoupling protein-1 downregulation in visceral fat. LLC-cachectic mice displayed FFA1 upregulation in subcutaneous fat, but not in visceral fat or gastrocnemius muscle, whereas FFA4 was unaltered. Overall, the present study shed new light on FFA1 and FFA4 receptors' role in metabolic disorders, indicating FFA1 receptor agonism as a promising strategy in mitigating cancer cachexia.


Subject(s)
Body Weight/drug effects , Cachexia/drug therapy , Carcinoma, Lewis Lung/metabolism , Docosahexaenoic Acids/therapeutic use , Receptors, G-Protein-Coupled/metabolism , alpha-Linolenic Acid/therapeutic use , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cachexia/etiology , Cachexia/metabolism , Carcinoma, Lewis Lung/complications , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Methylamines/pharmacology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Neoplasm Transplantation , Phenylpropionates/pharmacology , Propionates/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Sulfonamides/pharmacology , Xanthenes/pharmacology , alpha-Linolenic Acid/pharmacology
10.
Neurobiol Learn Mem ; 171: 107207, 2020 05.
Article in English | MEDLINE | ID: mdl-32147586

ABSTRACT

BACKGROUND AND PURPOSE: Hypoxia and cerebral ischemia (HI) events are capable of triggering important changes in brain metabolism, including glucose metabolism abnormalities, which may be related to the severity of the insult. Using positron emission microtomography (microPET) with [18F]fluorodeoxyglucose (18F-FDG), this study proposes to assess abnormalities of brain glucose metabolism in adult rats previously submitted to the neonatal HI model. We hypothesize that cerebral metabolic outcomes will be associated with cognitive deficits and magnitude of brain injury. METHODS: Seven-day-old rats were subjected to an HI model, induced by permanent occlusion of the right common carotid artery and systemic hypoxia. 18F-FDG-microPET was used to assess regional and whole brain glucose metabolism in rats at 60 postnatal days (PND 60). An interregional cross-correlation matrix was utilized to construct metabolic brain networks (MBN). Rats were also subjected to the Morris Water Maze (MWM) to evaluate spatial memory and their brains were processed for volumetric evaluation. RESULTS: Brain glucose metabolism changes were observed in adult rats after neonatal HI insult, limited to the right brain hemisphere. However, not all HI animals exhibited significant cerebral hypometabolism. Hippocampal glucose metabolism was used to stratify HI animals into HI hypometabolic (HI-h) and HI non-hypometabolic (HI non-h) groups. The HI-h group had drastic MBN disturbance, cognitive deficit, and brain tissue loss, concomitantly. Conversely, HI non-h rats had normal brain glucose metabolism and brain tissue preserved, but also presented MBN changes and spatial memory impairment. Furthermore, data showed that brain glucose metabolism correlated with cognitive deficits and brain volume outcomes. CONCLUSIONS: Our findings demonstrated that long-term changes in MBN drive memory impairments in adult rats subjected to neonatal hypoxic ischemia, using in vivo imaging microPET-FDG. The MBN analyses identified glucose metabolism abnormalities in HI non-h animals, which were not detected by conventional 18F-FDG standardized uptake value (SUVr) measurements. These animals exhibited a metabolic brain signature that may explain the cognitive deficit even with no identifiable brain damage.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Memory Disorders/metabolism , Nerve Net/metabolism , Animals , Brain/diagnostic imaging , Disease Models, Animal , Glucose/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/diagnostic imaging , Male , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Nerve Net/diagnostic imaging , Positron-Emission Tomography , Rats , Rats, Wistar
11.
Brain Struct Funct ; 224(8): 2857-2870, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31440907

ABSTRACT

Imaging studies have shown abnormal amygdala function in patients with posttraumatic stress disorder (PTSD). In addition, alterations in synaptic plasticity have been associated with psychiatric disorders and previous reports have indicated alterations in the amygdala morphology, especially in basolateral (BLA) neurons, are associated with stress-related disorders. Since, some individuals exposed to a traumatic event develop PTSD, the goals of this study were to evaluate the early effects of PTSD on amygdala glucose metabolism and analyze the possible BLA dendritic spine plasticity in animals with different levels of behavioral response. We employed the inescapable footshock protocol as an experimental model of PTSD and the animals were classified according to the duration of their freezing behavior into distinct groups: "extreme behavioral response" (EBR) and "minimal behavioral response". We evaluated the amygdala glucose metabolism at baseline (before the stress protocol) and immediately after the situational reminder using the microPET and the radiopharmaceutical 18F-FDG. The BLA dendritic spines were analyzed according to their number, density, shape and morphometric parameters. Our results show the EBR animals exhibited longer freezing behavior and increased proximal dendritic spines density in the BLA neurons. Neither the amygdaloid glucose metabolism, the types of dendritic spines nor their morphometric parameters showed statistically significant differences. The extreme behavior response induced by this PTSD protocol produces an early increase in BLA spine density, which is unassociated with either additional changes in the shape of spines or metabolic changes in the whole amygdala of Wistar rats.


Subject(s)
Basolateral Nuclear Complex/physiopathology , Dendritic Spines/physiology , Stress Disorders, Post-Traumatic/physiopathology , Animals , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/pathology , Dendritic Spines/pathology , Disease Models, Animal , Fluorodeoxyglucose F18 , Glucose/metabolism , Male , Positron-Emission Tomography , Rats, Wistar , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/pathology
12.
Brain Res ; 1722: 146355, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31356782

ABSTRACT

Stress has been considered as a risk factor for the development and aggravation of several diseases. The hypothalamic-pituitary-adrenal axis (HPA) is one of the main actors for the stress response and homeostasis maintenance. Positron emission tomography (PET) has been used to evaluate neuronal activity and to study brain regions that may be related to the HPA axis response. Since neuroimaging is an important tool in detecting neuroendocrine-related changes, we used fluorodeoxyglucose-18 (18F-FDG) and positron emission microtomography (microPET) to evaluate sexual differences in the glucose brain metabolism after 10, 30 and 40 min of acute stress in Balb/c mice. We also investigated the effects of restraint stress in blood, liver and adrenal gland 18F-FDG biodistribution using a gamma counter. A decreased glucose uptake in the whole brain in both females and males was found. Additionally, there were time and sex-dependent alterations in the 18F-FDG uptake after restraint stress in specific brain regions, indicating that males could be more vulnerable to the short-term effects of acute stress. According to the gamma counter biodistribution, only females showed a significant decreased glucose uptake in the blood, liver and right adrenal after restraint stress. In addition, in comparisons between the sexes, males showed a decreased glucose uptake in the whole brain and in several brain regions compared to females. In conclusion, exposure to acute restraint stress resulted in significant decreased glucose metabolism in the brain, with particular effects in different regions and organs in a sex-specific manner.


Subject(s)
Brain/metabolism , Glucose/metabolism , Sex Characteristics , Stress, Psychological/metabolism , Animals , Female , Fluorodeoxyglucose F18 , Male , Mice, Inbred BALB C , Positron-Emission Tomography , Restraint, Physical
13.
Brain Behav Immun ; 80: 879-888, 2019 08.
Article in English | MEDLINE | ID: mdl-31176000

ABSTRACT

Sepsis is characterized by a severe and disseminated inflammation. In the central nervous system, sepsis promotes synaptic dysfunction and permanent cognitive impairment. Besides sepsis-induced neuronal dysfunction, glial cell response has been gaining considerable attention with microglial activation as a key player. By contrast, astrocytes' role during acute sepsis is still underexplored. Astrocytes are specialized immunocompetent cells involved in brain surveillance. In this context, the potential communication between the peripheral immune system and astrocytes during acute sepsis still remains unclear. We hypothesized that peripheral blood mononuclear cell (PBMC) mediators are able to affect the brain during an episode of acute sepsis. With this in mind, we first performed a data-driven transcriptome analysis of blood from septic patients to identify common features among independent clinical studies. Our findings evidenced pronounced impairment in energy-related signaling pathways in the blood of septic patients. Since astrocytes are key for brain energy homeostasis, we decided to investigate the communication between PBMC mediators and astrocytes in a rat model of acute sepsis, induced by cecal ligation and perforation (CLP). In the CLP animals, we identified widespread in vivo brain glucose hypometabolism. Ex vivo analyses demonstrated astrocyte reactivity along with reduced glutamate uptake capacity during sepsis. Also, by exposing cultured astrocytes to mediators released by PBMCs from CLP animals, we reproduced the energetic failure observed in vivo. Finally, by pharmacologically inhibiting phosphoinositide 3-kinase (PI3K), a central metabolic pathway downregulated in the blood of septic patients and reduced in the CLP rat brain, we mimicked the PBMC mediators effect on glutamate uptake but not on glucose metabolism. These results suggest that PBMC mediators are capable of directly mediating astrocyte reactivity and contribute to the brain energetic failure observed in acute sepsis. Moreover, the evidence of PI3K participation in this process indicates a potential target for therapeutic modulation.


Subject(s)
Astrocytes/metabolism , Leukocytes, Mononuclear/metabolism , Sepsis/physiopathology , Adult , Animals , Brain/metabolism , Central Nervous System/metabolism , Databases, Genetic , Disease Models, Animal , Female , Glutamic Acid/metabolism , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/physiology , Male , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar , Sepsis/genetics , Signal Transduction/physiology
14.
Pain ; 160(6): 1383-1401, 2019 06.
Article in English | MEDLINE | ID: mdl-30720581

ABSTRACT

Generalized pain and fatigue are both hallmarks of fibromyalgia, a syndrome with an indefinite etiology. The treatment options for fibromyalgia are currently limited, probably because of its intricate pathophysiology. Thus, further basic and clinical research on this condition is currently needed. This study investigated the effects of nociceptin/orphanin FQ (N/OFQ) receptor (NOPr) ligands and the modulation of the NOP system in the preclinical mouse model of reserpine-induced fibromyalgia. The effects of administration of the natural agonist N/OFQ and the selective NOPr antagonists (UFP-101 and SB-612111) were evaluated in fibromyalgia-related symptoms in reserpine-treated mice. The expression of prepronociceptin/orphanin FQ and NOPr was assessed in central and peripheral sites at different time points after reserpine administration. Nociceptin/orphanin FQ displayed dual effects in the behavioral changes in the reserpine-elicited fibromyalgia model. The peptide NOPr antagonist UFP-101 produced analgesic and antifatigue effects, by preventing alterations in brain activity and skeletal muscle metabolism, secondary to fibromyalgia induction. The nonpeptide NOPr antagonist SB-612111 mirrored the favorable effects of UFP-101 in painful and fatigue alterations induced by reserpine. A time-related up- or downregulation of prepronociceptin/orphanin FQ and NOPr was observed in supraspinal, spinal, and peripheral sites of reserpine-treated mice. Our data shed new lights on the mechanisms underlying the fibromyalgia pathogenesis, supporting a role for N/OFQ-NOP receptor system in this syndrome.


Subject(s)
Analgesics/pharmacology , Fatigue/drug therapy , Fibromyalgia/drug therapy , Opioid Peptides/pharmacology , Animals , Disease Models, Animal , Female , Male , Mice , Narcotic Antagonists/pharmacology , Pain/drug therapy , Protein Precursors/pharmacology , Receptors, Opioid/drug effects , Nociceptin Receptor , Nociceptin
15.
Mol Neurobiol ; 55(12): 9307-9327, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29667130

ABSTRACT

Voltage-gated calcium channels (VGCCs) play a critical role in neuroinflammatory diseases, such as multiple sclerosis (MS). CTK 01512-2 is a recombinant version of the peptide Phα1ß derived from the spider Phoneutria nigriventer, which inhibits N-type VGCC/TRPA1-mediated calcium influx. We investigated the effects of this molecule in the mouse model of experimental autoimmune encephalomyelitis (EAE). The effects of CTK 01512-2 were compared to those displayed by ziconotide-a selective N-type VGCC blocker clinically used for chronic pain-and fingolimod-a drug employed for MS treatment. The intrathecal (i.t.) treatment with CTK 01512-2 displayed beneficial effects, by preventing nociception, body weight loss, splenomegaly, MS-like clinical and neurological scores, impaired motor coordination, and memory deficits, with an efficacy comparable to that observed for ziconotide and fingolimod. This molecule displayed a favorable profile on EAE-induced neuroinflammatory changes, including inflammatory infiltrate, demyelination, pro-inflammatory cytokine production, glial activation, and glucose metabolism in the brain and spinal cord. The recovery of spatial memory, besides a reduction of serum leptin levels, allied to central and peripheral elevation of the anti-inflammatory cytokine IL-10, was solely modulated by CTK 01512-2, dosed intrathecally. The intravenous (i.v.) administration of CTK 01512-2 also reduced the EAE-elicited MS-like symptoms, similarly to that seen in animals that received fingolimod orally. Ziconotide lacked any significant effect when dosed by i.v. route. Our results indicate that CTK 01512-2 greatly improved the neuroinflammatory responses in a mouse model of MS, with a higher efficacy when compared to ziconotide, pointing out this molecule as a promising adjuvant for MS management.


Subject(s)
Calcium Channel Blockers/therapeutic use , Multiple Sclerosis/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Calcium Channel Blockers/pharmacology , Chemokines/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Inflammation/pathology , Inflammation Mediators/metabolism , Injections, Spinal , Mice, Inbred C57BL , Motor Activity/drug effects , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Nociception/drug effects , Peptide Fragments/metabolism , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
16.
Epilepsia ; 59(5): 923-934, 2018 05.
Article in English | MEDLINE | ID: mdl-29600825

ABSTRACT

OBJECTIVE: Temporal lobe epilepsy (TLE) is one of the most common types of epilepsy syndromes in the world. Depression is an important comorbidity of epilepsy, which has been reported in patients with TLE and in different experimental models of epilepsy. However, there is no established consensus on which brain regions are associated with the manifestation of depression in epilepsy. Here, we investigated the alterations in cerebral glucose metabolism and the metabolic network in the pilocarpine-induced rat model of epilepsy and correlated it with depressive behavior during the chronic phase of epilepsy. METHODS: Fluorodeoxyglucose (18 F-FDG) was used to investigate the cerebral metabolism, and a cross-correlation matrix was used to examine the metabolic network in chronically epileptic rats using micro-positron emission tomography (microPET) imaging. An experimental model of epilepsy was induced by pilocarpine injection (320 mg/kg, ip). Forced swim test (FST), sucrose preference test (SPT), and eating-related depression test (ERDT) were used to evaluate depression-like behavior. RESULTS: Our results show an association between epilepsy and depression comorbidity based on changes in both cerebral glucose metabolism and the functional metabolic network. In addition, we have identified a significant correlation between brain glucose hypometabolism and depressive-like behavior in chronically epileptic rats. Furthermore, we found that the epileptic depressed group presents a hypersynchronous brain metabolic network in relation to the epileptic nondepressed group. SIGNIFICANCE: This study revealed relevant alterations in glucose metabolism and the metabolic network among the brain regions of interest for both epilepsy and depression pathologies. Thus it seems that depression in epileptic animals is associated with a more diffuse hypometabolism and altered metabolic network architecture and plays an important role in chronic epilepsy.


Subject(s)
Brain/metabolism , Depression/etiology , Epilepsy/metabolism , Epilepsy/psychology , Glucose/metabolism , Animals , Brain/physiopathology , Comorbidity , Depression/metabolism , Epilepsy/physiopathology , Image Interpretation, Computer-Assisted , Male , Positron-Emission Tomography , Rats , Rats, Wistar
17.
Mol Neurobiol ; 55(3): 2025-2041, 2018 03.
Article in English | MEDLINE | ID: mdl-28271402

ABSTRACT

This study was performed to evaluate the bilateral effects of focal permanent ischemia (FPI) on glial metabolism in the cerebral cortex. Two and 9 days after FPI induction, we analyze [18F]FDG metabolism by micro-PET, astrocyte morphology and reactivity by immunohistochemistry, cytokines and trophic factors by ELISA, glutamate transporters by RT-PCR, monocarboxylate transporters (MCTs) by western blot, and substrate uptake and oxidation by ex vivo slices model. The FPI was induced surgically by thermocoagulation of the blood in the pial vessels of the motor and sensorimotor cortices in adult (90 days old) male Wistar rats. Neurochemical analyses were performed separately on both ipsilateral and contralateral cortical hemispheres. In both cortical hemispheres, we observed an increase in tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), and glutamate transporter 1 (GLT-1) mRNA levels; lactate oxidation; and glutamate uptake and a decrease in brain-derived neurotrophic factor (BDNF) after 2 days of FPI. Nine days after FPI, we observed an increase in TNF-α levels and a decrease in BDNF, GLT-1, and glutamate aspartate transporter (GLAST) mRNA levels in both hemispheres. Additionally, most of the unilateral alterations were found only in the ipsilateral hemisphere and persisted until 9 days post-FPI. They include diminished in vivo glucose uptake and GLAST expression, followed by increased glial fibrillary acidic protein (GFAP) gray values, astrocyte reactivity, and glutamate oxidation. Astrocytes presented signs of long-lasting reactivity, showing a radial morphology. In the intact hemisphere, there was a decrease in MCT2 levels, which did not persist. Our study shows the bilateralism of glial modifications following FPI, highlighting the role of energy metabolism adaptations on brain recovery post-ischemia.


Subject(s)
Adaptation, Physiological/physiology , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Neuroglia/metabolism , Animals , Brain Ischemia/pathology , Cerebral Cortex/pathology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Male , Neuroglia/pathology , Rats , Rats, Wistar
18.
CNS Neurosci Ther ; 21(5): 463-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25645708

ABSTRACT

AIMS: In previous studies, transplantation of bone marrow mononuclear cells (BMMCs) in epileptic animals has been found to be neuroprotective. However, the mechanism by which the BMMCs act remains unclear. We hypothesize that BMMCs may provide neuroprotection to the epileptic brain through trophic support. To test our hypothesis, we studied the temporal expression of neurotrophins after BMMC transplantation in the epileptic rat hippocampus. METHODS: Chronically epileptic rats were intravenously transplanted with 1 × 10(7) BMMCs isolated from GFP transgenic mice. Expression levels of BDNF, GDNF, NGF, VEGF, and TGF-ß1, and their receptors, were evaluated by ELISA and/or qRT-PCR analysis. RESULTS: Our data revealed increased protein expression of BDNF, GDNF, NGF, and VEGF and reduced levels of TGF-ß1 in the hippocampus of transplanted epileptic animals. Additionally, an increase in the mRNA expression of BDNF, GDNF, and VEGF, a reduction in TGF-ß1, and a decrease in mRNA levels of the TrkA and TGFR-ß1 receptors were also observed. CONCLUSION: The gain provided by transplanted BMMCs in the epileptic brain may be related to the ability of these cells in modulating the network of neurotrophins and angiogenic signals.


Subject(s)
Bone Marrow Transplantation , Epilepsy/metabolism , Epilepsy/therapy , Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Bone Marrow Cells/metabolism , Chronic Disease , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Pilocarpine , Rats, Wistar
19.
Coluna/Columna ; 12(4): 274-277, 2013. ilus, graf
Article in English | LILACS | ID: lil-699028

ABSTRACT

OBJECTIVE: We studied transplants of bone marrow mononuclear cells (BMMC) by lumbar puncture (LP) in a severe model of spinal cord injury (SCI) using clip compression. METHODS: BMMCs or saline solution were transplanted by LP 48 hours and 9 days post injury. Motor function was evaluated by BBB scale, histological analysis by Nissl technique and the verification of cell migration by PCR analysis. RESULTS: The BBB had significantly improved in rats treated with BMMCs by LP compared with controls (p<0.001). The histological analysis did not showed difference in the lesional area between the groups. The PCR analysis was able to found BMMCs in the injury site. CONCLUSIONS: two BMMC transplants by LP improved motor function in a severe model of SCI and BMMC was found in the injury site.


OBJETIVO: Estudamos transplantes de células mononucleares da medula óssea (CMMO) por punção lombar (PL) em um modelo de lesão da medula espinal (LME) grave usando compressão por clipe. MÉTODOS: CMMOs ou solução salina foram transplantadas por PL 48 horas e 9 dias após a LME. A função motora foi avaliada pela escala de BBB, a análise histológica pela técnica de Nissl e a migração celular pela análise de reação em cadeia da polimerase (PCR). RESULTADOS: A BBB demonstrou melhora significativa nos animais tratados com CMMOs por PL, em comparação com os controles (p < 0,001). A análise histológica não apresentou diferença entre as áreas de lesão dos grupos. CMMOs foram encontradas no local da lesão na análise de PCR. CONCLUSÃO: Dois transplantes de CMMOs por PL melhoraram a função motora em um modelo de LME grave. CMMOs foram encontradas no local da lesão.


OBJETIVO: Estudiamos trasplantes de células mononucleares de la médula ósea (CMMO) por punción lumbar (PL) en un modelo de lesión de la médula espinal (LME) grave, usando compresión por clip. MÉTODOS: CMMOs o solución salina fueron trasplantadas por PL, 48 horas y 9 días después de la LME. La función motora fue evaluada por la escala de BBB, el análisis histológico por la técnica de Nissl y la migración celular por el análisis de reacción en cadena de la polimerasa (PCR). RESULTADOS: La BBB demostró mejoría significativa en los animales tratados con CMMOs por PL, en comparación con los controles (p < 0,001). El análisis histológico no presentó diferencia entre las áreas de lesión de los grupos. CMMOs fueron encontradas en el lugar de la lesión en el análisis de PCR. CONCLUSIÓN: Dos trasplantes de CMMOs por PL mejoraron la función motora en un modelo de LME grave. CMMOs fueron encontradas en el área de la lesión.


Subject(s)
Humans , Bone Marrow Transplantation , Spinal Cord Injuries , Spinal Puncture , Bone Marrow Cells
20.
Life Sci ; 89(7-8): 229-34, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21718708

ABSTRACT

AIMS: Epilepsy affects 0.5-1% of the world's population, and approximately a third of these patients are refractory to current medication. Given their ability to proliferate, differentiate and regenerate tissues, stem cells could restore neural circuits lost during the course of the disease and reestablish the physiological excitability of neurons. This study verified the therapeutic potential of bone marrow mononuclear cells (BMMCs) on seizure control and cognitive impairment caused by experimentally induced epilepsy. MAIN METHODS: Status epilepticus (SE) was induced by lithium-pilocarpine injection and controlled with diazepam 90 min after SE onset. Lithium-pilocarpine-treated rats were intravenously transplanted 22 days after SE with BMMCs obtained from enhanced green fluorescent protein (eGFP) transgenic C57BL/6 mice. Control epileptic animals were given an equivalent volume of saline or fibroblast injections. Animals were video-monitored for the presence of spontaneous recurrent seizures prior to and following the cell administration procedure. In addition, rats underwent cognitive evaluation using a Morris water maze. KEY FINDINGS: Our data show that BMMCs reduced the frequency of seizures and improved the learning and long-term spatial memory impairments of epileptic rats. EGFP-positive cells were detected in the brains of transplanted animals by PCR analysis. SIGNIFICANCE: The positive behavioral effects observed in our study indicate that BMMCs could represent a promising therapeutic option in the management of chronic temporal lobe epilepsy.


Subject(s)
Cell Transplantation , Cognition Disorders/prevention & control , Epilepsy/therapy , Memory Disorders/prevention & control , Monocytes/cytology , Seizures/prevention & control , Animals , Bone Marrow Cells/cytology , Chronic Disease , Cognition Disorders/chemically induced , Diazepam/pharmacology , Disease Models, Animal , Epilepsy/complications , Lithium/pharmacology , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Monocytes/transplantation , Pilocarpine/pharmacology , Rats , Rats, Wistar , Seizures/chemically induced , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL