Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Gut Microbes ; 15(2): 2249146, 2023 12.
Article in English | MEDLINE | ID: mdl-37668317

ABSTRACT

Long-term sequelae of coronavirus disease (COVID)-19 are frequent and of major concern. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection affects the host gut microbiota, which is linked to disease severity in patients with COVID-19. Here, we report that the gut microbiota of post-COVID subjects had a remarkable predominance of Enterobacteriaceae strains with an antibiotic-resistant phenotype compared to healthy controls. Additionally, short-chain fatty acid (SCFA) levels were reduced in feces. Fecal transplantation from post-COVID subjects to germ-free mice led to lung inflammation and worse outcomes during pulmonary infection by multidrug-resistant Klebsiella pneumoniae. transplanted mice also exhibited poor cognitive performance. Overall, we show prolonged impacts of SARS-CoV-2 infection on the gut microbiota that persist after subjects have cleared the virus. Together, these data demonstrate that the gut microbiota can directly contribute to post-COVID sequelae, suggesting that it may be a potential therapeutic target.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Animals , Mice , SARS-CoV-2 , Anti-Bacterial Agents , Disease Progression
2.
Neurosci Lett ; 792: 136955, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36347339

ABSTRACT

GPR139 is an orphan G-protein-coupled receptor that is expressed in restricted areas of the nervous system, including the hypothalamus. In this study, we hypothesized that GPR139 could be involved in the regulation of energy balance and metabolism. In the first part of the study, we confirmed that GPR139 is expressed in the hypothalamus and particularly in proopiomelanocortin and agouti-related peptide neurons of the mediobasal hypothalamus. Using a lentivirus with a short-hairpin RNA, we inhibited the expression of GPR139 bilaterally in the mediobasal hypothalamus of mice. The intervention promoted a 40% reduction in the hypothalamic expression of GPR139, which was accompanied by an increase in body mass, a reduction in fasting blood glucose levels, and an increase in insulin levels. In the hypothalamus, inhibition of GPR139 was accompanied by a reduction in the expression of orexin. As previous studies using a pharmacological antagonist of orexin showed a beneficial impact on type 2 diabetes and glucose metabolism, we propose that the inhibition of hypothalamic GPR139 could be acting indirectly through the orexin system to control systemic glucose and insulin. In conclusion, this study advances the characterization of GPR139 in the hypothalamus, demonstrating its involvement in the regulation of body mass, blood insulin, and glycemia.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin , Mice , Animals , Orexins/metabolism , Insulin/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Receptors, G-Protein-Coupled/metabolism , Nerve Tissue Proteins/metabolism
3.
Cells ; 11(13)2022 06 22.
Article in English | MEDLINE | ID: mdl-35805082

ABSTRACT

BACKGROUND: The hypothalamic proopiomelanocortin (Pomc) neurons act as first-order sensors of systemic energy stores, providing signals that regulate caloric intake and energy expenditure. In experimental obesity, dietary saturated fatty acids affect Pomc endopeptidases (PCs), resulting in the abnormal production of the neurotransmitters α-melanocyte-stimulating hormone (α-MSH) and ß-endorphin, thus impacting energy balance. The cAMP response element-binding protein (CREB) is one of the transcription factors that control the expression of Pomc endopeptidases; however, it was previously unknown if dietary fats could affect CREB and consequently the expression of Pomc endopeptidases. METHODS: Here, we used single-cell RNA sequencing analysis, PCR, immunoblot, ELISA and immunofluorescence histological assays to determine the impact of a high-fat diet (HFD) on the expression and function of hypothalamic CREB and its impact on the melanocortinergic system. RESULTS: The results indicate that CREB is expressed in arcuate nucleus Pomc neurons and is activated as early as nine hours after the introduction of a high-fat diet. The inhibition of hypothalamic CREB using a short-hairpin RNA lentiviral vector resulted in increased diet-induced body-mass gain and reduced energy expenditure. This was accompanied by reduced expression of the Pomc endopeptidases, protein convertase 2, which are encoded by Pcsk2, and by the loss of the high-fat-diet-induced effect to inhibit the production of α-MSH. CONCLUSIONS: This study provides the first evidence for the involvement of CREB in the abnormal regulation of the hypothalamic Pomc endopeptidase system in experimental obesity.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Pro-Opiomelanocortin , Diet, High-Fat , Endopeptidases , Humans , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Proprotein Convertase 2 , alpha-MSH/pharmacology
4.
Neuropharmacology ; 208: 108983, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35143850

ABSTRACT

In experimental models, hypothalamic dysfunction is a key component of the pathophysiology of diet-induced obesity. Early after the introduction of a high-fat diet, neurons, microglia, astrocytes and tanycytes of the mediobasal hypothalamus undergo structural and functional changes that impact caloric intake, energy expenditure and systemic glucose tolerance. Inflammation has emerged as a central component of this response, and as in other inflammatory conditions, there is a time course of events that determine the fate of distinct cells involved in the central regulation of whole-body energy homeostasis. Here, we review the work that identified key mechanisms, cellular players and temporal features of diet-induced hypothalamic abnormalities. This article is part of the special Issue on 'Cross Talk between Periphery and the Brain'.


Subject(s)
Hypothalamus , Obesity , Diet, High-Fat , Energy Metabolism/physiology , Humans , Neuroglia , Neurons
5.
Front Neurosci ; 15: 734158, 2021.
Article in English | MEDLINE | ID: mdl-34803583

ABSTRACT

Worldwide, and especially in Western civilizations, most of the staple diets contain high amounts of fat and refined carbohydrates, leading to an increasing number of obese individuals. In addition to inducing metabolic disorders, energy dense food intake has been suggested to impair brain functions such as cognition and mood control. Here we demonstrate an impaired memory function already 3 days after the start of a high-fat diet (HFD) exposure, and depressive-like behavior, in the tail suspension test, after 5 days. These changes were followed by reduced synaptic density, changes in mitochondrial function and astrocyte activation in the hippocampus. Preceding or coinciding with the behavioral changes, we found an induction of the proinflammatory cytokines TNF-α and IL-6 and an increased permeability of the blood-brain barrier (BBB), in the hippocampus. Finally, in mice treated with a TNF-α inhibitor, the behavioral and BBB alterations caused by HFD-feeding were mitigated suggesting that inflammatory signaling was critical for the changes. In summary, our findings suggest that HFD rapidly triggers hippocampal dysfunction associated with BBB disruption and neuroinflammation, promoting a progressive breakdown of synaptic and metabolic function. In addition to elucidating the link between diet and cognitive function, our results might be relevant for the comprehension of the neurodegenerative process.

6.
Metab Brain Dis ; 36(8): 2425-2436, 2021 12.
Article in English | MEDLINE | ID: mdl-34599738

ABSTRACT

The search for strategies to develop resilience against metabolic and neuropsychiatric disorders has motivated the clinical and experimental assessment of early life interventions such as lifestyle-based and use of unconventional pharmacological compounds. In this study, we assessed the effects of voluntary physical activity and 7,8-Dihydroxy-4-methylcoumarin (DHMC), independently or in combination, over mice physiological and behavioral parameters, adult hippocampal and hypothalamic neurogenesis, and neurotrophic factors expression in the hypothalamus. C57Bl/6J mice were submitted to a 29-day treatment with DHMC and allowed free access to a running wheel. We found that DHMC treatment alone reduced fasting blood glucose levels. Moreover, physical activity showed an anxiolytic effect in the elevated plus maze task and DHMC produced additional anxiolytic behavior, evidenced by reduced activity during the light cycle in the physical activity group. Although we did not find any differences in hypothalamic or hippocampal adult neurogenesis, DHMC increased gene expression levels of VEGF, which was correlated to the reduced fasting glucose levels. In conclusion, our data emphasize the potential of physical activity in reducing development of neuropsychiatric conditions, such as anxiety, and highlights DHMC as an attractive compound to be investigated in future studies addressing neuropsychiatric disorders associated with metabolic conditions.


Subject(s)
Coumarins , Neuronal Plasticity , Animals , Coumarins/pharmacology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL
7.
J Neuroinflammation ; 18(1): 192, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34465367

ABSTRACT

BACKGROUND: Interleukin-6 (IL6) produced in the context of exercise acts in the hypothalamus reducing obesity-associated inflammation and restoring the control of food intake and energy expenditure. In the hippocampus, some of the beneficial actions of IL6 are attributed to its neurogenesis-inducing properties. However, in the hypothalamus, the putative neurogenic actions of IL6 have never been explored, and its potential to balance energy intake can be an approach to prevent or attenuate obesity. METHODS: Wild-type (WT) and IL6 knockout (KO) mice were employed to study the capacity of IL6 to induce neurogenesis. We used cell labeling with Bromodeoxyuridine (BrdU), immunofluorescence, and real-time PCR to determine the expression of markers of neurogenesis and neurotransmitters. We prepared hypothalamic neuroprogenitor cells from KO that were treated with IL6 in order to provide an ex vivo model to further characterizing the neurogenic actions of IL6 through differentiation assays. In addition, we analyzed single-cell RNA sequencing data and determined the expression of IL6 and IL6 receptor in specific cell types of the murine hypothalamus. RESULTS: IL6 expression in the hypothalamus is low and restricted to microglia and tanycytes, whereas IL6 receptor is expressed in microglia, ependymocytes, endothelial cells, and astrocytes. Exogenous IL6 reduces diet-induced obesity. In outbred mice, obesity-resistance is accompanied by increased expression of IL6 in the hypothalamus. IL6 induces neurogenesis-related gene expression in the hypothalamus and in neuroprogenitor cells, both from WT as well as from KO mice. CONCLUSION: IL6 induces neurogenesis-related gene expression in the hypothalamus of WT mice. In KO mice, the neurogenic actions of IL6 are preserved; however, the appearance of new fully differentiated proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons is either delayed or disturbed.


Subject(s)
Hypothalamus/metabolism , Interleukin-6/genetics , Neurogenesis/genetics , Neurons/metabolism , Obesity/genetics , Animals , Energy Metabolism/physiology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Hypothalamus/drug effects , Interleukin-6/metabolism , Interleukin-6/pharmacology , Male , Mice , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Obesity/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
8.
Sci Rep ; 11(1): 18114, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518585

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. While cognitive deficits remain the major manifestation of AD, metabolic and non-cognitive abnormalities, such as alterations in food intake, body weight and energy balance are also present, both in AD patients and animal models. In this sense, the tauroursodeoxycholic acid (TUDCA) has shown beneficial effects both in reducing the central and cognitive markers of AD, as well as in attenuating the metabolic disorders associated with it. We previously demonstrated that TUDCA improves glucose homeostasis and decreases the main AD neuromarkers in the streptozotocin-induced AD mouse model (Stz). Besides that, TUDCA-treated Stz mice showed lower body weight and adiposity. Here, we investigated the actions of TUDCA involved in the regulation of body weight and adiposity in Stz mice, since the effects of TUDCA in hypothalamic appetite control and energy homeostasis have not yet been explored in an AD mice model. The TUDCA-treated mice (Stz + TUDCA) displayed lower food intake, higher energy expenditure (EE) and respiratory quotient. In addition, we observed in the hypothalamus of the Stz + TUDCA mice reduced fluorescence and gene expression of inflammatory markers, as well as normalization of the orexigenic neuropeptides AgRP and NPY expression. Moreover, leptin-induced p-JAK2 and p-STAT3 signaling in the hypothalamus of Stz + TUDCA mice was improved, accompanied by reduced acute food intake after leptin stimulation. Taken together, we demonstrate that TUDCA treatment restores energy metabolism in Stz mice, a phenomenon that is associated with reduced food intake, increased EE and improved hypothalamic leptin signaling. These findings suggest treatment with TUDCA as a promising therapeutic intervention for the control of energy homeostasis in AD individuals.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Energy Metabolism/drug effects , Homeostasis , Streptozocin/adverse effects , Taurochenodeoxycholic Acid/pharmacology , Adiposity , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Biomarkers , Body Weight , Disease Management , Disease Models, Animal , Gene Expression , Immunohistochemistry , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Leptin/metabolism , Male , Mice , Organ Specificity , Signal Transduction , Thermogenesis
9.
Sci Rep ; 11(1): 15453, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326383

ABSTRACT

Glutamic acid is the main excitatory neurotransmitter acting both in the brain and in peripheral tissues. Abnormal distribution of glutamic acid receptors occurs in skin hyperproliferative conditions such as psoriasis and skin regeneration; however, the biological function of glutamic acid in the skin remains unclear. Using ex vivo, in vivo and in silico approaches, we showed that exogenous glutamic acid promotes hair growth and keratinocyte proliferation. Topical application of glutamic acid decreased the expression of genes related to apoptosis in the skin, whereas glutamic acid increased cell viability and proliferation in human keratinocyte cultures. In addition, we identified the keratinocyte glutamic acid excitotoxic concentration, providing evidence for the existence of a novel skin signalling pathway mediated by a neurotransmitter that controls keratinocyte and hair follicle proliferation. Thus, glutamic acid emerges as a component of the peripheral nervous system that acts to control cell growth in the skin. These results raise the perspective of the pharmacological and nutritional use of glutamic acid to treat skin diseases.


Subject(s)
Glutamic Acid/pharmacology , Hair Follicle/drug effects , Hair/drug effects , Skin Physiological Phenomena , Skin/drug effects , Animals , Apoptosis , Cell Line , Cell Proliferation , Computer Simulation , Drug Development , Fibroblasts/metabolism , Glutamic Acid/metabolism , Humans , Keratinocytes/cytology , Male , Mice , Protein Interaction Mapping , Regeneration , Signal Transduction , Skin/metabolism
10.
Mol Cell Endocrinol ; 521: 111116, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33321116

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder and the major cause of dementia. According to predictions of the World Health Organization, more than 150 million people worldwide will suffer from dementia by 2050. An increasing number of studies have associated AD with type 2 diabetes mellitus (T2DM), since most of the features found in T2DM are also observed in AD, such as insulin resistance and glucose intolerance. In this sense, some bile acids have emerged as new therapeutic targets to treat AD and metabolic disorders. The taurine conjugated bile acid, tauroursodeoxycholic (TUDCA), reduces amyloid oligomer accumulation and improves cognition in APP/PS1 mice model of AD, and also improves glucose-insulin homeostasis in obese and type 2 diabetic mice. Herein, we investigated the effect of TUDCA upon glucose metabolism in streptozotocin-induced AD mice model (Stz). The Stz mice that received 300 mg/kg TUDCA during 10 days (Stz + TUDCA), showed improvement in glucose tolerance and insulin sensitivity, reduced fasted and fed glycemia, increased islet mass and ß-cell area, as well as increased glucose-stimulated insulin secretion, compared with Stz mice that received only PBS. Stz + TUDCA mice also displayed lower neuroinflammation, reduced protein content of amyloid oligomer in the hippocampus, improved memory test and increased protein content of insulin receptor ß-subunit in the hippocampus. In conclusion, TUDCA treatment enhanced glucose homeostasis in the streptozotocin-induced Alzheimer's disease mice model, pointing this bile acid as a good strategy to counteract glucose homeostasis disturbance in AD pathology.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Bile Acids and Salts/metabolism , Blood Glucose/drug effects , Hippocampus/drug effects , Insulin-Secreting Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Glucose/metabolism , Glucose/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/drug therapy , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Memory and Learning Tests , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Streptozocin/toxicity , Taurochenodeoxycholic Acid/administration & dosage
11.
Nutr Neurosci ; 24(12): 978-988, 2021 Dec.
Article in English | MEDLINE | ID: mdl-31910791

ABSTRACT

Although the benefits of moderate intake of red wine in decreasing incidence of cardiovascular diseases associated to hypercholesterolemia are well recognized, there are still widespread misconceptions about its effects on the hypercholesterolemia-related cognitive impairments. Herein we investigated the putative benefits of regular red wine consumption on cognitive performance of low-density lipoprotein receptor knockout (LDLr-/-) mice, an animal model of familial hypercholesterolemia, which display cognitive impairments since early ages. The red wine was diluted into the drinking water to a final concentration of 6% ethanol and was available for 60 days for LDLr-/- mice fed a normal or high-cholesterol diet. The results indicated that moderate red wine consumption did not alter locomotor parameters and liver toxicity. Across multiple cognitive tasks evaluating spatial learning/reference memory and recognition/identification memory, hypercholesterolemic mice drinking red wine performed significantly better than water group, regardless of diet. Additionally, immunofluorescence assays indicated a reduction of astrocyte activation and lectin stain in the hippocampus of LDLr-/- mice under consumption of red wine. These findings demonstrate that the moderate consumption of red wine attenuates short- and long-term memory decline associated with hypercholesterolemia in mice and suggest that it could be through a neurovascular action.


Subject(s)
Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Hypercholesterolemia/complications , Receptors, LDL/physiology , Wine , Animals , Behavior, Animal , Brain/blood supply , Cholesterol, Dietary/administration & dosage , Disease Models, Animal , Hippocampus/physiopathology , Hypercholesterolemia/genetics , Hypercholesterolemia/physiopathology , Liver Diseases, Alcoholic , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Receptors, LDL/deficiency , Receptors, LDL/genetics
12.
J Alzheimers Dis ; 78(1): 97-115, 2020.
Article in English | MEDLINE | ID: mdl-32925052

ABSTRACT

BACKGROUND: Evidence has revealed an association between familial hypercholesterolemia and cognitive impairment. In this regard, a connection between cognitive deficits and hippocampal blood-brain barrier (BBB) breakdown was found in low-density lipoprotein receptor knockout mice (LDLr-/-), a mouse model of familial hypercholesterolemia. OBJECTIVE: Herein we investigated the impact of a hypercholesterolemic diet on cognition and BBB function in C57BL/6 wild-type and LDLr-/-mice. METHODS: Animals were fed with normal or high cholesterol diets for 30 days. Thus, wild-type and LDLr-/-mice were submitted to memory paradigms. Additionally, BBB integrity was evaluated in the mice's prefrontal cortices and hippocampi. RESULTS: A tenfold elevation in plasma cholesterol levels of LDLr-/-mice was observed after a hypercholesterolemic diet, while in wild-type mice, the hypercholesterolemic diet exposure increased plasma cholesterol levels only moderately and did not induce cognitive impairment. LDLr-/-mice presented memory impairment regardless of the diet. We observed BBB disruption as an increased permeability to sodium fluorescein in the prefrontal cortices and hippocampi and a decrease on hippocampal claudin-5 and occludin mRNA levels in both wild-type and LDLr-/-mice treated with a hypercholesterolemic diet. The LDLr-/-mice fed with a regular diet already presented BBB dysfunction. The BBB-increased leakage in the hippocampi of LDLr-/-mice was related to high microvessel content and intense astrogliosis, which did not occur in the control mice. CONCLUSION: Therefore, LDLr-/-mice seem to be more susceptible to cognitive impairments and BBB damage induced by exposure to a high cholesterol diet. Finally, BBB disruption appears to be a relevant event in hypercholesterolemia-induced brain alterations.


Subject(s)
Blood-Brain Barrier , Cholesterol/metabolism , Cognitive Dysfunction/metabolism , Hypercholesterolemia/metabolism , Animals , Cognition , Diet , Disease Models, Animal , Gliosis/metabolism , Hippocampus/metabolism , Male , Memory , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Prefrontal Cortex/metabolism , Receptors, LDL
13.
Sci Rep ; 10(1): 11047, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632088

ABSTRACT

Hypothalamic adult neurogenesis provides the basis for renewal of neurons involved in the regulation of whole-body energy status. In addition to hormones, cytokines and growth factors, components of the diet, particularly fatty acids, have been shown to stimulate hypothalamic neurogenesis; however, the mechanisms behind this action are unknown. Here, we hypothesized that GPR40 (FFAR1), the receptor for medium and long chain unsaturated fatty acids, could mediate at least part of the neurogenic activity in the hypothalamus. We show that a GPR40 ligand increased hypothalamic cell proliferation and survival in adult mice. In postnatal generated neurospheres, acting in synergy with brain-derived neurotrophic factor (BDNF) and interleukin 6, GPR40 activation increased the expression of doublecortin during the early differentiation phase and of the mature neuronal marker, microtubule-associated protein 2 (MAP2), during the late differentiation phase. In Neuro-2a proliferative cell-line GPR40 activation increased BDNF expression and p38 activation. The chemical inhibition of p38 abolished GPR40 effect in inducing neurogenesis markers in neurospheres, whereas BDNF immunoneutralization inhibited GPR40-induced cell proliferation in the hypothalamus of adult mice. Thus, GPR40 acts through p38 and BDNF to induce hypothalamic neurogenesis. This study provides mechanistic advance in the understating of how a fatty acid receptor regulates adult hypothalamic neurogenesis.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Neurogenesis/physiology , Receptors, G-Protein-Coupled/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Hypothalamus/drug effects , Imidazoles/pharmacology , Interleukin-6/physiology , Ligands , Male , Methylamines/pharmacology , Mice , Mice, Inbred C57BL , Models, Neurological , Neurons/drug effects , Neurons/physiology , Propionates/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
Neuroscience ; 440: 210-229, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32497756

ABSTRACT

NAD(P)+ transhydrogenase (NNT) links redox states of the mitochondrial NAD(H) and NADP(H) via a reaction coupled to proton-motive force across the inner mitochondrial membrane. NNT is believed to be ubiquitously present in mammalian cells, but its expression may vary substantially in different tissues. The present study investigated the tissue distribution and possible roles of NNT in the mouse brain. The pons exhibited high NNT expression/activity, and immunohistochemistry revealed intense NNT labeling in neurons from brainstem nuclei. In some of these regions, neuronal NNT labeling was strongly colocalized with enzymes involved in the biosynthesis of 5-hydroxytryptamine (5-HT) and nitric oxide (NO), which directly or indirectly require NADPH. Behavioral tests were performed in mice lacking NNT activity (Nnt-/-, mice carrying the mutated NntC57BL/6J allele from the C57BL/6J strain) and the Nnt+/+ controls. Our data demonstrated that aged Nnt-/- mice (18-20 months old), but not adult mice (3-4 months old), showed an increased immobility time in the tail suspension test that was reversed by fluoxetine treatment, providing evidence of depressive-like behavior in these mice. Aged Nnt-/- mice also exhibited behavioral changes and impaired locomotor activity in the open field and rotarod tests. Despite the colocalization between NNT and NO synthase, the S-nitrosation and cGMP levels were independent of the Nnt genotype. Taken together, our results indicated that NNT is unevenly distributed throughout the brain and associated with 5-THergic and NOergic neurons. The lack of NNT led to alterations in brain functions related to mood and motor behavior/performance in aged mice.


Subject(s)
NADP Transhydrogenase, AB-Specific , NAD , Animals , Brain/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , NADP/metabolism , NADP Transhydrogenase, AB-Specific/metabolism
15.
Pharmacol Rep ; 72(3): 563-570, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32240535

ABSTRACT

BACKGROUND: Experimental and clinical studies indicate that neuronal death with the presence of high levels of reactive oxygen species are present in depressed patients and antidepressants might display neuroprotective effects against them. However, the mechanisms underlying antidepressant neuroprotection are not completely understood. In our previous study, we showed that mirtazapine modulated the expression of pro- and anti-apoptotic proteins in mouse brain structures, but there are no data in human cells. Thus, this work was designed to study the possible neuroprotective properties of mirtazapine and imipramine, two commercially available antidepressants with different primary mechanisms of action, in human neuroblastoma SH-SY5Y cells against an oxidative insult. METHODS: SH-SY5Y cells were preincubated with mirtazapine and imipramine (1-20 µM) for 24 h, then hydrogen peroxide (H2O2) was added into the medium containing the antidepressants for additional 24 h, and MTT assay was carried out subsequently. Also, to elucidate the molecular mechanism underlying the neuroprotective properties of antidepressants, we investigated the effects of mirtazapine and imipramine (2 µM) in pro- and anti-apoptotic proteins gene expression in SH-SY5Y cells. RESULTS: Mirtazapine (1 and 2 µM) and imipramine (1and 2 µM) protected against hydrogen peroxide-induced cellular viability impairment. Most importantly, both compounds reduced p53 mRNA expression, but only imipramine enhanced the Bcl-2/Bax ratio. CONCLUSIONS: The obtained data indicate that mirtazapine and imipramine have neuroprotective effects against H2O2-induced cell death. Although both antidepressants reduced Bax and p53 mRNA expression, only the protection mediated by imipramine might be due to its ability to enhance Bcl-2/Bax ratio.


Subject(s)
Apoptosis/drug effects , Imipramine/pharmacology , Mirtazapine/pharmacology , Neuroprotective Agents/pharmacology , Cell Line, Tumor , Cell Survival , Humans , Neuroblastoma/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-Associated Death Protein/metabolism
16.
J Alzheimers Dis ; 73(2): 585-596, 2020.
Article in English | MEDLINE | ID: mdl-31815695

ABSTRACT

Familial hypercholesterolemia (FH) is a genetic disorder caused by dysfunction of low density lipoprotein receptors (LDLr), resulting in elevated plasma cholesterol levels. FH patients frequently exhibit cognitive impairment, a finding recapitulated in LDLr deficient mice (LDLr-/-), an animal model of FH. In addition, LDLr-/- mice are more vulnerable to the deleterious memory impact of amyloid-ß (Aß), a peptide linked to Alzheimer's disease. Here, we investigated whether the expression of proteins involved in Aß metabolism are altered in the brains of adult or middle-aged LDLr-/- mice. After spatial memory assessment, Aß levels and gene expression of LDLr related-protein 1, proteins involved in Aß synthesis, and apoptosis-related proteins were evaluated in prefrontal cortex and hippocampus. Moreover, the location and cell-specificity of apoptosis signals were evaluated. LDLr-/- mice presented memory impairment, which was more severe in middle-aged animals. Memory deficit in LDLr-/- mice was not associated with altered expression of proteins involved in Aß processing or changes in Aß levels in either hippocampus or prefrontal cortex. We further found that the expression of Bcl-2 was reduced while the expression of Bax was increased in both prefrontal cortex and hippocampus in 3- and 14-month-old LDLr-/-mice Finally, LDLr-/- mice presented increased immunoreactivity for activated caspase-3 in the prefrontal cortex and hippocampus. The activation of caspase 3 was predominantly associated with neurons in LDLr-/- mice. Cognitive impairment in LDLr-/- mice is thus accompanied by an exacerbation of neuronal apoptosis in brain regions related to memory formation, but not by changes in Aß processing or levels.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Apoptosis/genetics , Brain Chemistry/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Aging/metabolism , Aging/psychology , Animals , Caspase 3 , Cholesterol/blood , Gene Expression , Hippocampus/metabolism , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Prefrontal Cortex/metabolism
17.
Brain Behav Immun ; 87: 272-285, 2020 07.
Article in English | MEDLINE | ID: mdl-31863824

ABSTRACT

Interleukin-17 (IL-17) is expressed in the intestine in response to changes in the gut microbiome landscape and plays an important role in intestinal and systemic inflammatory diseases. There is evidence that dietary factors can also modify the expression of intestinal IL-17. Here, we hypothesized that, similar to several other gut-produced factors, IL-17 may act in the hypothalamus to modulate food intake. We confirm that food intake increases IL-17 expression in the mouse ileum and human blood. There is no expression of IL-17 in the hypothalamus; however, IL-17 receptor A is expressed in both pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons. Upon systemic injection, IL-17 promoted a rapid increase in hypothalamic POMC expression, which was followed by a late increase in the expression of AgRP. Both systemic and intracerebroventricular injections of IL-17 reduced calorie intake without affecting whole-body energy expenditure. Systemic but not intracerebroventricular injection of IL-17 increase brown adipose tissue temperature. Thus, IL-17 is a gut-produced factor that is controlled by diet and modulates food intake by acting in the hypothalamus. Our findings provide the first evidence of a cytokine that is acutely regulated by food intake and plays a role in the regulation of eating.


Subject(s)
Hypothalamus , Interleukin-17 , Agouti-Related Protein/metabolism , Animals , Eating , Humans , Hypothalamus/metabolism , Mice , Pro-Opiomelanocortin/metabolism
18.
Mol Metab ; 30: 1-15, 2019 12.
Article in English | MEDLINE | ID: mdl-31767163

ABSTRACT

OBJECTIVE: In familial hypercholesterolemia (FH), mutations in the low-density lipoprotein (LDL) receptor (LDLr) gene result in increased plasma LDL cholesterol. Clinical and preclinical studies have revealed an association between FH and hippocampus-related memory and mood impairment. We here asked whether hippocampal pathology in FH might be a consequence of compromised adult hippocampal neurogenesis. METHODS: We evaluated hippocampus-dependent behavior and neurogenesis in adult C57BL/6JRj and LDLr-/- mice. We investigated the effects of elevated cholesterol and the function of LDLr in neural precursor cells (NPC) isolated from adult C57BL/6JRj mice in vitro. RESULTS: Behavioral tests revealed that adult LDLr-/- mice showed reduced performance in a dentate gyrus (DG)-dependent metric change task. This phenotype was accompanied by a reduction in cell proliferation and adult neurogenesis in the DG of LDLr-/- mice, suggesting a potential direct impact of LDLr mutation on NPC. Exposure of NPC to LDL as well as LDLr gene knockdown reduced proliferation and disrupted transcriptional activity of genes involved in endogenous cholesterol synthesis and metabolism. The LDL treatment also induced an increase in intracellular lipid storage. Functional analysis of differentially expressed genes revealed parallel modulation of distinct regulatory networks upon LDL treatment and LDLr knockdown. CONCLUSIONS: Together, these results suggest that high LDL levels and a loss of LDLr function, which are characteristic to individuals with FH, might contribute to a disease-related impairment in adult hippocampal neurogenesis and, consequently, cognitive functions.


Subject(s)
Hippocampus/metabolism , Hyperlipoproteinemia Type II/metabolism , Receptors, LDL/metabolism , Animals , Cholesterol/metabolism , Cholesterol, LDL/blood , Hypercholesterolemia , Hyperlipoproteinemia Type II/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neural Stem Cells/metabolism , Neurogenesis/physiology , Phenotype , Receptors, LDL/genetics
19.
Redox Biol ; 20: 118-129, 2019 01.
Article in English | MEDLINE | ID: mdl-30308475

ABSTRACT

Oxidative stress and mitochondrial dysfunction are critical events in neurodegenerative diseases; therefore, molecules that increase cellular antioxidant defenses represent a future pharmacologic strategy to counteract such conditions. The aim of this study was to investigate the potential protective effect of (PhSe)2 on mouse hippocampal cell line (HT22) exposed to tert-BuOOH (in vitro model of oxidative stress), as well as to elucidate potential mechanisms underlying this protection. Our results showed that tert-BuOOH caused time- and concentration-dependent cytotoxicity, which was preceded by increased oxidants production and mitochondrial dysfunction. (PhSe)2 pre-incubation significantly prevented these cytotoxic events and the observed protective effects were paralleled by the upregulation of the cellular glutathione-dependent antioxidant system: (PhSe)2 increased GSH levels (> 60%), GPx activity (6.9-fold) and the mRNA expression of antioxidant enzymes Gpx1 (3.9-fold) and Gclc (2.3-fold). Of note, the cytoprotective effect of (PhSe)2 was significantly decreased when cells were treated with mercaptosuccinic acid, an inhibitor of GPx, indicating the involvement of GPx modulation in the observed protective effect. In summary, the present findings bring out a new action mechanism concerning the antioxidant properties of (PhSe)2. The observed upregulation of the glutathione-dependent antioxidant system represents a future pharmacologic possibility that goes beyond the well-known thiol-peroxidase activity of this compound.


Subject(s)
Benzene Derivatives/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Glutathione/metabolism , Lipid Peroxidation/drug effects , Mice , Models, Biological , Oxidants/biosynthesis , Oxidation-Reduction/drug effects
20.
Mol Neurobiol ; 55(1): 435-444, 2018 01.
Article in English | MEDLINE | ID: mdl-27966074

ABSTRACT

Sepsis survivors frequently develop late cognitive impairment. Because little is known on the mechanisms of post-septic memory deficits, there are no current effective approaches to prevent or treat such symptoms. Here, we subjected mice to severe sepsis induced by cecal ligation and puncture (CLP) and evaluated the sepsis-surviving animals in the open field, novel object recognition (NOR), and step-down inhibitory avoidance (IA) task at different times after surgery. Post-septic mice (30 days post-surgery) failed in the NOR and IA tests but exhibited normal performance when re-evaluated 45 days after surgery. Cognitive impairment in post-septic mice was accompanied by reduced hippocampal levels of proteins involved in synaptic plasticity, including synaptophysin, cAMP response element-binding protein (CREB), CREB phosphorylated at serine residue 133 (CREBpSer133), and GluA1 phosphorylated at serine residue 845 (GluA1pSer845). Expression of tumor necrosis factor α (TNF-α) was increased and brain insulin signaling was disrupted, as indicated by increased hippocampal IRS-1 phosphorylation at serine 636 (IRS-1pSer636) and decreased phosphorylation of IRS-1 at tyrosine 465 (IRS-1pTyr465), in the hippocampus 30 days after CLP. Phosphorylation of Akt at serine 473 (AktpSer473) and of GSK3 at serine 9 (GSK3ßpSer9) were also decreased in hippocampi of post-septic animals, further indicating that brain insulin signaling is disrupted by sepsis. We then treated post-septic mice with liraglutide, a GLP-1 receptor agonist with insulinotropic activity, or TDZD-8, a GSK3ß inhibitor, which rescued NOR memory. In conclusion, these results establish that hippocampal inflammation and disrupted insulin signaling are induced by sepsis and are linked to late memory impairment in sepsis survivors.


Subject(s)
Brain/metabolism , Cognitive Dysfunction/metabolism , Insulin/metabolism , Sepsis/metabolism , Signal Transduction/physiology , Animals , Brain/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Exploratory Behavior/physiology , Male , Mice , Sepsis/complications , Sepsis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...