Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Eur J Endocrinol ; 188(3): R37-R45, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36883605

ABSTRACT

Obesity and its comorbidities are long-standing, challenging global health problems. Lack of exercise, overnutrition, and especially the consumption of fat-rich foods are some of the most important factors leading to an increase in prevalence in modern society. The pathophysiology of obesity as a metabolic inflammatory disease has moved into focus since new therapeutic approaches are required. The hypothalamus, a brain area responsible for energy homeostasis, has recently received special attention in this regard. Hypothalamic inflammation was identified to be associated with diet-induced obesity and new evidence suggests that it may be, beyond that, a pathological mechanism of the disease. This inflammation impairs the local signaling of insulin and leptin leading to dysfunction of the regulation of energy balance and thus, weight gain. After a high-fat diet consumption, activation of inflammatory mediators such as the nuclear factor κB or c-Jun N-terminal kinase pathway can be observed, accompanied by elevated secretion of pro-inflammatory interleukins and cytokines. Brain resident glia cells, especially microglia and astrocytes, initiate this release in response to the flux of fatty acids. The gliosis occurs rapidly before the actual weight gain. Dysregulated hypothalamic circuits change the interaction between neuronal and non-neuronal cells, contributing to the establishment of inflammatory processes. Several studies have reported reactive gliosis in obese humans. Although there is evidence for a causative role of hypothalamic inflammation in the obesity development, data on underlying molecular pathways in humans are limited. This review discusses the current state of knowledge on the relationship between hypothalamic inflammation and obesity in humans.


Subject(s)
Gliosis , Obesity , Humans , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Obesity/metabolism , Hypothalamus/metabolism , Weight Gain , Inflammation , Diet, High-Fat , Energy Metabolism
2.
Endocr Rev ; 44(2): 281-296, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36251886

ABSTRACT

Accumulated preclinical literature demonstrates that hypothalamic inflammation and gliosis are underlying causal components of diet-induced obesity in rodent models. This review summarizes and synthesizes available translational data to better understand the applicability of preclinical findings to human obesity and its comorbidities. The published literature in humans includes histopathologic analyses performed postmortem and in vivo neuroimaging studies measuring indirect markers of hypothalamic tissue microstructure. Both support the presence of hypothalamic inflammation and gliosis in children and adults with obesity. Findings predominantly point to tissue changes in the region of the arcuate nucleus of the hypothalamus, although findings of altered tissue characteristics in whole hypothalamus or other hypothalamic regions also emerged. Moreover, the severity of hypothalamic inflammation and gliosis has been related to comorbid conditions, including glucose intolerance, insulin resistance, type 2 diabetes, and low testosterone levels in men, independent of elevated body adiposity. Cross-sectional findings are augmented by a small number of prospective studies suggesting that a greater degree of hypothalamic inflammation and gliosis may predict adiposity gain and worsening insulin sensitivity in susceptible individuals. In conclusion, existing human studies corroborate a large preclinical literature demonstrating that hypothalamic neuroinflammatory responses play a role in obesity pathogenesis. Extensive or permanent hypothalamic tissue remodeling may negatively affect the function of neuroendocrine regulatory circuits and promote the development and maintenance of elevated body weight in obesity and/or comorbid endocrine disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Male , Adult , Child , Humans , Gliosis/etiology , Gliosis/pathology , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Prospective Studies , Hypothalamus , Obesity/complications , Inflammation
3.
FASEB J ; 35(10): e21869, 2021 10.
Article in English | MEDLINE | ID: mdl-34469026

ABSTRACT

The leucine-rich repeat-containing family 8 member A (LRRC8A) is an essential subunit of the volume-regulated anion channel (VRAC). VRAC is critical for cell volume control, but its broader physiological functions remain under investigation. Recent studies in the field indicate that Lrrc8a disruption in the brain astrocytes reduces neuronal excitability, impairs synaptic plasticity and memory, and protects against cerebral ischemia. In the present work, we generated brain-wide conditional LRRC8A knockout mice (LRRC8A bKO) using NestinCre -driven Lrrc8aflox/flox excision in neurons, astrocytes, and oligodendroglia. LRRC8A bKO animals were born close to the expected Mendelian ratio and developed without overt histological abnormalities, but, surprisingly, all died between 5 and 9 weeks of age with a seizure phenotype, which was confirmed by video and EEG recordings. Brain slice electrophysiology detected changes in the excitability of pyramidal cells and modified GABAergic inputs in the hippocampal CA1 region of LRRC8A bKO. LRRC8A-null hippocampi showed increased immunoreactivity of the astrocytic marker GFAP, indicating reactive astrogliosis. We also found decreased whole-brain protein levels of the GABA transporter GAT-1, the glutamate transporter GLT-1, and the astrocytic enzyme glutamine synthetase. Complementary HPLC assays identified reduction in the tissue levels of the glutamate and GABA precursor glutamine. Together, these findings suggest that VRAC provides vital control of brain excitability in mouse adolescence. VRAC deletion leads to a lethal phenotype involving progressive astrogliosis and dysregulation of astrocytic uptake and supply of amino acid neurotransmitters and their precursors.


Subject(s)
Astrocytes/pathology , Gliosis/mortality , Glutamic Acid/metabolism , Membrane Proteins/physiology , Seizures/mortality , Animals , Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Female , Gliosis/etiology , Gliosis/pathology , Ion Transport , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Seizures/etiology , Seizures/pathology
4.
Sci Rep ; 11(1): 12613, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131204

ABSTRACT

Secondary injury following cortical stroke includes delayed gliosis and eventual neuronal loss in the thalamus. However, the effects of aging and the potential to ameliorate this gliosis with NMDA receptor (NMDAR) antagonism are not established. We used the permanent distal middle cerebral artery stroke model (pdMCAO) to examine secondary thalamic injury in young and aged mice. At 3 days post-stroke (PSD3), slight microgliosis (IBA-1) and astrogliosis (GFAP) was evident in thalamus, but no infarct. Gliosis increased dramatically through PSD14, at which point degenerating neurons were detected. Flow cytometry demonstrated a significant increase in CD11b+/CD45int microglia (MG) in the ipsilateral thalamus at PSD14. CCR2-RFP reporter mouse further demonstrated that influx of peripheral monocytes contributed to the MG/Mϕ population. Aged mice demonstrated reduced microgliosis and astrogliosis compared with young mice. Interestingly, astrogliosis demonstrated glial scar-like characteristics at two years post-stroke, but not by 6 weeks. Lastly, treatment with memantine (NMDAR antagonist) at 4 and 24 h after stroke significantly reduced gliosis at PSD14. These findings expand our understanding of gliosis in the thalamus following cortical stroke and demonstrate age-dependency of this secondary injury. Additionally, these findings indicate that delayed treatment with memantine (an FDA approved drug) provides significant reduction in thalamic gliosis.


Subject(s)
Gliosis/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Memantine/pharmacology , Stroke/drug therapy , Aging/drug effects , Aging/pathology , Animals , Brain Ischemia/complications , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Disease Models, Animal , Gliosis/etiology , Gliosis/pathology , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Mice , Neuroprotective Agents/pharmacology , Stroke/complications , Thalamus/drug effects , Thalamus/pathology
5.
Molecules ; 26(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806413

ABSTRACT

MsrB1 used to be named selenoprotein R, for it was first identified as a selenocysteine containing protein by searching for the selenocysteine insert sequence (SECIS) in the human genome. Later, it was found that MsrB1 is homologous to PilB in Neisseria gonorrhoeae, which is a methionine sulfoxide reductase (Msr), specifically reducing L-methionine sulfoxide (L-Met-O) in proteins. In humans and mice, four members constitute the Msr family, which are MsrA, MsrB1, MsrB2, and MsrB3. MsrA can reduce free or protein-containing L-Met-O (S), whereas MsrBs can only function on the L-Met-O (R) epimer in proteins. Though there are isomerases existent that could transfer L-Met-O (S) to L-Met-O (R) and vice-versa, the loss of Msr individually results in different phenotypes in mice models. These observations indicate that the function of one Msr cannot be totally complemented by another. Among the mammalian Msrs, MsrB1 is the only selenocysteine-containing protein, and we recently found that loss of MsrB1 perturbs the synaptic plasticity in mice, along with the astrogliosis in their brains. In this review, we summarized the effects resulting from Msr deficiency and the bioactivity of selenium in the central nervous system, especially those that we learned from the MsrB1 knockout mouse model. We hope it will be helpful in better understanding how the trace element selenium participates in the reduction of L-Met-O and becomes involved in neurobiology.


Subject(s)
Central Nervous System/pathology , Gliosis/pathology , Methionine Sulfoxide Reductases/physiology , Neuronal Plasticity , Selenium/metabolism , Animals , Central Nervous System/metabolism , Gliosis/etiology , Gliosis/metabolism , Humans , Mice , Mice, Knockout
6.
Cell Tissue Res ; 383(2): 677-692, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32960358

ABSTRACT

Peripheral nerve injury is associated with spinal microgliosis which plays a pivotal role in the development of neuropathic pain behavior. Several agents of primary afferent origin causing the microglial reaction have been identified, but the type(s) of primary afferents that release these mediators are still unclear. In this study, specific labeling of C-fiber spinal afferents by lectin histochemistry and selective chemodenervation by capsaicin were applied to identify the type(s) of primary afferents involved in the microglial response. Comparative quantitative morphometric evaluation of the microglial reaction in central projection territories of intact and injured peripheral nerves in the superficial (laminae I and II) and deep (laminae III and IV) spinal dorsal horn revealed a significant, about three-fold increase in microglial density after transection of the sciatic or the saphenous nerve. Prior perineural treatment of these nerves with capsaicin, resulting in a selective defunctionalization of C-fiber afferent fibers failed to affect spinal microgliosis. Similarly, peripheral nerve injury-induced increase in microglial density was unaffected in rats treated neonatally with capsaicin known to result in a near-total loss of C-fiber dorsal root fibers. Perineural treatment with capsaicin per se did not evoke a significant increase in microglial density. These observations indicate that injury-induced spinal microgliosis may be attributed to phenotypic changes in injured myelinated primary afferent neurons, whereas the contribution of C-fiber primary sensory neurons to this neuroimmune response is negligible. Spinal myelinated primary afferents may play a hitherto unrecognized role in regulation of neuroimmune and perisynaptic microenvironments of the spinal dorsal horn.


Subject(s)
Capsaicin/therapeutic use , Gliosis/drug therapy , Gliosis/etiology , Peripheral Nerve Injuries/complications , Spinal Cord/pathology , Animals , Animals, Newborn , Capsaicin/pharmacology , Cell Count , Gliosis/pathology , Male , Peripheral Nerve Injuries/pathology , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Rats, Wistar , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology
7.
Horm Behav ; 120: 104675, 2020 04.
Article in English | MEDLINE | ID: mdl-31923417

ABSTRACT

Consumption of a high fat diet (HFD) increases circulating free fatty acids, which can enter the brain and promote a state of microgliosis, as defined by a change in microglia number and/or morphology. Most studies investigating diet-induced microgliosis have been conducted in male rodents despite well-documented sex differences in the neural control of food intake and neuroimmune signaling. This highlights the need to investigate how sex hormones may modulate the behavioral and cellular response to HFD consumption. Estradiol is of particular interest since it exerts a potent anorexigenic effect and has both anti-inflammatory and neuroprotective effects in the brain. As such, the aim of the current study was to investigate whether estradiol attenuates the development of HFD-induced microgliosis in female rats. Estradiol- and vehicle-treated ovariectomized rats were fed either a low-fat chow diet or a 60% HFD for 4 days, after which they were perfused and brain sections were processed via immunohistochemistry for microglia-specific Iba1 protein. Four days of HFD consumption promoted microgliosis, as measured via an increase in the number of microglia in the arcuate nucleus (ARC) of the hypothalamus and nucleus of the solitary tract (NTS), and a decrease in microglial branching in the ARC, NTS, lateral hypothalamus (LH), and ventromedial hypothalamus. Estradiol replacement attenuated the HFD-induced changes in microglia accumulation and morphology in the ARC, LH, and NTS. We conclude that estradiol has protective effects against HFD-induced microgliosis in a region-specific manner in hypothalamic and hindbrain areas implicated in the neural control of food intake.


Subject(s)
Diet, High-Fat/adverse effects , Estradiol/pharmacology , Gliosis/prevention & control , Microglia/drug effects , Ovariectomy/adverse effects , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/pathology , Brain Diseases/etiology , Brain Diseases/pathology , Brain Diseases/prevention & control , Cell Count , Cell Size/drug effects , Dietary Fats/adverse effects , Estradiol/deficiency , Female , Gliosis/etiology , Gliosis/pathology , Hypothalamus/metabolism , Hypothalamus/pathology , Microglia/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Solitary Nucleus/drug effects , Solitary Nucleus/pathology
8.
Mol Neurobiol ; 57(5): 2194-2205, 2020 May.
Article in English | MEDLINE | ID: mdl-31974940

ABSTRACT

Hypoxic-ischemic (HI) brain injury remains an important cause of brain damage in neonates with potential life-long consequences. Caffeine, which is a competitive inhibitor of adenosine receptors, is commonly used as treatment for preterm apnoea in clinical settings. In the current study, we investigated the effects of caffeine given at 0 h, 6 h, 12 h or 24 h after HI in P10 mouse pups. Open field and rotarod behavioural tests were performed 2 weeks after injury, and brain morphology was then evaluated. Gene expression and immunohistological analyses were assessed in mice 1- and 5-day post-HI. A single dose of caffeine directly after HI resulted in a reduction of the lesion in the grey and white matter, judged by immunostaining of MAP2 and MBP, respectively, compared to PBS-treated controls. In addition, the number of amoeboid microglia and apoptotic cells, the area covered by astrogliosis, and the expression of pro-inflammatory cytokines were significantly decreased. Behavioural assessment after 2 weeks showed increased open-field activity after HI, and this was normalised if caffeine was administered immediately after the injury. Later administrations of caffeine did not change the outcomes when compared to the vehicle group. In conclusion, caffeine only yielded neuroprotection and immunomodulation in a neonatal model of brain hypoxia ischaemia if administered immediately after injury.


Subject(s)
Caffeine/administration & dosage , Hypoxia-Ischemia, Brain/drug therapy , Immunomodulation/drug effects , Neuroglia/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/administration & dosage , Purinergic P1 Receptor Antagonists/administration & dosage , Animals , Brain Injuries/pathology , Caffeine/pharmacology , Caffeine/therapeutic use , DNA Fragmentation/drug effects , Demyelinating Diseases/prevention & control , Drug Administration Schedule , Drug Evaluation, Preclinical , Exploratory Behavior , Female , Gliosis/etiology , Gliosis/prevention & control , Hypoxia-Ischemia, Brain/metabolism , Inflammation/genetics , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Neuroglia/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Purinergic P1 Receptor Antagonists/pharmacology , Purinergic P1 Receptor Antagonists/therapeutic use , Random Allocation , Rotarod Performance Test , Single-Blind Method , Specific Pathogen-Free Organisms
9.
Horm Behav ; 120: 104690, 2020 04.
Article in English | MEDLINE | ID: mdl-31954709

ABSTRACT

Changes to neonatal nutrition result in long-lasting impairments in energy balance, which may be described as metabolic programing. Astrocytes, which are interconnected by gap junctions, have emerged as important players in the hypothalamic control of food intake. In order to study the effects of nutritional programming on glial morphology and protein expression, cross-fostered male Wistar rats at postnatal day 3 were assigned to three groups based on litter size: small litter (3 pups per dam, SL), normal litter (10 pups per dam, NL), and large litter (16 pups per dam, LL). Rats from the SL group exhibited higher body weight throughout the study and hyperphagia after weaning. LL animals exhibited hyperphagia, high energy efficiency and catch-up of body weight after weaning. Both the SL and LL groups at postnatal day 60 (PN60) exhibited increased levels of plasma leptin, the Lee index (as an index of obesity), adiposity content, immunoreactivity toward T-cell protein tyrosine phosphatase (TCPTP), and glial fibrillary acidic protein (GFAP) in the arcuate nucleus (ARC) of the hypothalamus. Astrocyte morphology was altered in the ARC of SL and LL animals, and this effect occurred in parallel with a reduction in immunoreactivity toward connexin 30 (CX30). The data obtained demonstrate that both neonatal over- and underfeeding promote not only alterations in the metabolic status but also morphological changes in glial cells in parallel with increasing TCPTP and changes in connexin expression.


Subject(s)
Animal Nutritional Physiological Phenomena , Connexins/genetics , Gliosis/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Adiposity/physiology , Animals , Animals, Newborn , Connexins/metabolism , Female , Gene Expression Regulation, Developmental , Gliosis/genetics , Gliosis/metabolism , Hyperphagia/complications , Hyperphagia/genetics , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamus/metabolism , Litter Size/physiology , Male , Obesity/complications , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Pregnancy , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Rats , Rats, Wistar , Sex Factors , Time Factors
10.
Mol Ther ; 27(8): 1507-1526, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31138511

ABSTRACT

Gliosis in Niemann-Pick type C (NP-C) disease is characterized by marked changes in microglia and astrocytes. However, the gliosis onset and progression in NP-C has not been systematically studied, nor has the mechanism underlying this finding. Here, we found early gliosis in the subventricular zone (SVZ) of NP-C mice. Neural progenitor damage by Npc1 mutation suppressed vascular endothelial growth factor (VEGF) expression and further induced microglia activation followed by astrogliosis. Interestingly, excessive astrogliosis in the SVZ induced neural progenitor retention and/or migration into thalamus via astrocyte-derived VEGF, resulting in acceleration of thalamic and cortical gliosis through thalamo-cortical pathways. Transplantation of VEGF-overexpressing neural stem cells into the SVZ improved whole-brain pathology of NP-C mice. Overall, our data provide a new pathological perspective on NP-C neural pathology, revealing abnormalities in the subventricular-thalamo-cortical circuit of NP-C mouse brain and highlighting the importance of the SVZ microenvironment as a therapeutic target for NP-C disease.


Subject(s)
Cerebral Cortex/metabolism , Lateral Ventricles/metabolism , Niemann-Pick Disease, Type C/metabolism , Signal Transduction , Thalamus/metabolism , Animals , Astrocytes/metabolism , Biomarkers , Cell Movement , Disease Models, Animal , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Mice , Microglia/metabolism , Neural Stem Cells/metabolism , Niemann-Pick Disease, Type C/etiology , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/therapy , Vascular Endothelial Growth Factor A/metabolism
11.
Exp Eye Res ; 169: 1-12, 2018 04.
Article in English | MEDLINE | ID: mdl-29355737

ABSTRACT

Photobiomodulation (PBM) with 670 nm light has been shown to accelerate wound healing in soft tissue injuries, and also to protect neuronal tissues. However, little data exist on its effects on the non-neuronal components of the retina, such as Müller cells (MCs), which are the principal macroglia of the retina that play a role in maintaining retinal homeostasis. The aim of this study was to explore the effects of 670 nm light on activated MCs using in vivo and in vitro stress models. Adult Sprague-Dawley rats were exposed to photo-oxidative damage (PD) for 24 h and treated with 670 nm light at 0, 3 and 14 days after PD. Tissue was collected at 30 days post-PD for analysis. Using the in vitro scratch model with a human MC line (MIO-M1), area coverage and cellular stress were analysed following treatment with 670 nm light. We showed that early treatment with 670 nm light after PD reduced MC activation, lowering the retinal expression of GFAP and FGF-2. 670 nm light treatment mitigated the production of MC-related pro-inflammatory cytokines (including IL-1ß), and reduced microglia/macrophage (MG/MΦ) recruitment into the outer retina following PD. This subsequently decreased photoreceptor loss, slowing the progression of retinal degeneration. In vitro, we showed that 670 nm light directly modulated MC activation, reducing rates of area coverage by suppressing cellular proliferation and spreading. This study indicates that 670 nm light treatment post-injury may have therapeutic benefit when administered shortly after retinal damage, and could be useful for retinal degenerations where MC gliosis is a feature of disease progression.


Subject(s)
Ependymoglial Cells/radiation effects , Gliosis/therapy , Phototherapy/methods , Radiation Injuries, Experimental/therapy , Radiation Injuries/therapy , Retina/radiation effects , Retinal Degeneration/therapy , Animals , Cell Line , Cell Movement , Cell Survival , Cytokines/metabolism , Disease Models, Animal , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Fibroblast Growth Factor 2/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Humans , Light/adverse effects , Oxidative Stress , Radiation Injuries/etiology , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retina/pathology , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
12.
Endocrinology ; 159(1): 368-387, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29077836

ABSTRACT

Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.


Subject(s)
Astrocytes/metabolism , Gliosis/etiology , Hyperphagia/physiopathology , Hypothalamic Diseases/etiology , Hypothalamus/metabolism , Microglia/metabolism , Adiposity , Age Factors , Animals , Animals, Newborn , Astrocytes/immunology , Astrocytes/pathology , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Female , Gene Expression Regulation, Developmental , Gliosis/immunology , Gliosis/metabolism , Gliosis/pathology , Hypothalamic Diseases/immunology , Hypothalamic Diseases/metabolism , Hypothalamic Diseases/pathology , Hypothalamus/immunology , Hypothalamus/pathology , Inflammation Mediators/metabolism , Male , Microglia/immunology , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats, Wistar , Sex Characteristics , Signal Transduction , Weight Gain
13.
J Neuroinflammation ; 14(1): 201, 2017 Oct 12.
Article in English | MEDLINE | ID: mdl-29025435

ABSTRACT

BACKGROUND: The epidemic of obesity has reached alarming levels in both developing and developed nations. Excessive calorie intake and sedentary lifestyle due to technological advancements are the main causal factors for overweight and obesity among the human population. Obesity has been associated with a number of co-morbidities such as hypertension, type 2 diabetes mellitus, cardiovascular diseases, and neurodegeneration and dementia. The progression of neurological disorders in obese subjects has been mainly attributed to neuroinflammation. Withania somnifera has been used in numerous Ayurvedic formulations owing to its wide array of health-promoting properties. The current study was designed to test the hypothesis whether dry leaf powder of W. somnifera has anxiolytic and anti-neuroinflammatory potential in diet-induced obesity. METHODS: Young adult female rats were divided into four groups: low fat diet group (LFD) fed with regular chow feed, high fat diet group (HFD) fed with diet containing 30% fat by weight, low fat diet plus extract group (LFDE) fed with regular chow feed supplemented with dry leaf powder of W. somnifera 1 mg/g of body weight (ASH), and high fat diet plus extract group (HFDE) fed with diet containing 30% fat by weight and supplemented with ASH. All the animals were kept on respective feeding regimen for 12 weeks; following which, the animals were tested for their anxiety-like behavior using elevated plus maze test. The animals were sacrificed and used to study various inflammatory markers such as GFAP, Iba1, PPARγ, iNOS, MCP-1, TNFα, IL-1ß, IL-6, and various markers of NF-κB pathway by Western blotting and quantitative real-time PCR. Serum levels of leptin, insulin and pro-inflammatory cytokines were also assayed. RESULTS: ASH treated rats showed less anxiety levels as compared to HFD animals. At molecular level, ASH ameliorated the HFD-induced reactive gliosis and microgliosis and suppressed the expression of inflammatory markers such as PPARγ, iNOS, MCP-1, TNFα, IL-1ß, and IL-6. Further, ASH ameliorated leptin and insulin resistance and prevented HFD-induced apoptosis. CONCLUSIONS: Dry leaf powder of W. somnifera may prove to be a potential therapeutic agent to attenuate neuroinflammation associated with obesity and may prevent its co-morbidities.


Subject(s)
Anxiety/drug therapy , Diet, High-Fat/adverse effects , Encephalitis/drug therapy , Plant Extracts/therapeutic use , Withania , Animals , Anxiety/blood , Anxiety/etiology , Apoptosis/drug effects , Body Weight/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Encephalitis/blood , Encephalitis/etiology , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Gliosis/drug therapy , Gliosis/etiology , Hyperinsulinism/drug therapy , Hyperinsulinism/etiology , Hyperlactatemia/drug therapy , Maze Learning/drug effects , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Rats , Signal Transduction/drug effects
14.
Mol Metab ; 6(8): 897-908, 2017 08.
Article in English | MEDLINE | ID: mdl-28752053

ABSTRACT

OBJECTIVE: The hypothalamus of hypercaloric diet-induced obese animals is featured by a significant increase of microglial reactivity and its associated cytokine production. However, the role of dietary components, in particular fat and carbohydrate, with respect to the hypothalamic inflammatory response and the consequent impact on hypothalamic control of energy homeostasis is yet not clear. METHODS: We dissected the different effects of high-carbohydrate high-fat (HCHF) diets and low-carbohydrate high-fat (LCHF) diets on hypothalamic inflammatory responses in neurons and non-neuronal cells and tested the hypothesis that HCHF diets induce hypothalamic inflammation via advanced glycation end-products (AGEs) using mice lacking advanced glycation end-products (AGEs) receptor (RAGE) and/or the activated leukocyte cell-adhesion molecule (ALCAM). RESULTS: We found that consumption of HCHF diets, but not of LCHF diets, increases microgliosis as well as the presence of N(ε)-(Carboxymethyl)-Lysine (CML), a major AGE, in POMC and NPY neurons of the arcuate nucleus. Neuron-secreted CML binds to both RAGE and ALCAM, which are expressed on endothelial cells, microglia, and pericytes. On a HCHF diet, mice lacking the RAGE and ALCAM genes displayed less microglial reactivity and less neovasculature formation in the hypothalamic ARC, and this was associated with significant improvements of metabolic disorders induced by the HCHF diet. CONCLUSIONS: Combined overconsumption of fat and sugar, but not the overconsumption of fat per se, leads to excessive CML production in hypothalamic neurons, which, in turn, stimulates hypothalamic inflammatory responses such as microgliosis and eventually leads to neuronal dysfunction in the control of energy metabolism.


Subject(s)
Dietary Fats/metabolism , Dietary Sugars/metabolism , Gliosis/metabolism , Hypothalamus/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Dietary Fats/adverse effects , Dietary Sugars/adverse effects , Gliosis/etiology , Glycation End Products, Advanced/metabolism , Hypothalamus/pathology , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics
15.
Neurobiol Dis ; 105: 257-270, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28610891

ABSTRACT

Mucolipidosis type IV (MLIV) is a lysosomal storage disease exhibiting progressive intellectual disability, motor impairment, and premature death. There is currently no cure or corrective treatment. The disease results from mutations in the gene encoding mucolipin-1, a transient receptor potential channel believed to play a key role in lysosomal calcium egress. Loss of mucolipin-1 and subsequent defects lead to a host of cellular aberrations, including accumulation of glycosphingolipids (GSLs) in neurons and other cell types, microgliosis and, as reported here, cerebellar Purkinje cell loss. Several studies have demonstrated that N-butyldeoxynojirimycin (NB-DNJ, also known as miglustat), an inhibitor of the enzyme glucosylceramide synthase (GCS), successfully delays the onset of motor deficits, improves longevity, and rescues some of the cerebellar abnormalities (e.g., Purkinje cell death) seen in another lysosomal disease known as Niemann-Pick type C (NPC). Given the similarities in pathology between MLIV and NPC, we examined whether miglustat would be efficacious in ameliorating disease progression in MLIV. Using a full mucolipin-1 knockout mouse (Mcoln1-/-), we found that early miglustat treatment delays the onset and progression of motor deficits, delays cerebellar Purkinje cell loss, and reduces cerebellar microgliosis characteristic of MLIV disease. Quantitative mass spectrometry analyses provided new data on the GSL profiles of murine MLIV brain tissue and showed that miglustat partially restored the wild type profile of white matter enriched lipids. Collectively, our findings indicate that early miglustat treatment delays the progression of clinically relevant pathology in an MLIV mouse model, and therefore supports consideration of miglustat as a therapeutic agent for MLIV disease in humans.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Cerebellum/pathology , Enzyme Inhibitors/therapeutic use , Gliosis/drug therapy , Movement Disorders/drug therapy , Mucolipidoses , Purkinje Cells/drug effects , 1-Deoxynojirimycin/therapeutic use , Animals , Antigens, CD/metabolism , Cell Count , Disease Models, Animal , Exploratory Behavior/drug effects , Gliosis/etiology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Movement Disorders/etiology , Mucolipidoses/complications , Mucolipidoses/genetics , Mucolipidoses/pathology , Nerve Tissue Proteins/metabolism , Psychomotor Performance/drug effects , Purkinje Cells/pathology , Retina/pathology , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
16.
Mol Cell Neurosci ; 80: 123-133, 2017 04.
Article in English | MEDLINE | ID: mdl-28286294

ABSTRACT

An important hallmark of various neurodegenerative disorders is the proliferation and activation of microglial cells, the resident immune cells of the central nervous system (CNS). Mice that lack multifunctional protein-2 (MFP2), the key enzyme in peroxisomal ß-oxidation, develop excessive microgliosis that positively correlates with behavioral deficits whereas no neuronal loss occurs. However, the precise contribution of neuroinflammation to the fatal neuropathology of MFP2 deficiency remains largely unknown. Here, we first attempted to suppress the inflammatory response by administering various anti-inflammatory drugs but they failed to reduce microgliosis. Subsequently, Mfp2-/- mice were treated with the selective colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 as microglial proliferation and survival is dependent on CSF1R signaling. This resulted in the elimination of >95% of microglia from control mice but only 70% of the expanded microglial population from Mfp2-/- mice. Despite microglial diminution in Mfp2-/- brain, inflammatory markers remained unaltered and residual microglia persisted in a reactive state. CSF1R inhibition did not prevent neuronal dysfunction, cognitive decline and clinical deterioration of Mfp2-/- mice. Collectively, the unaltered inflammatory profile despite suppressed microgliosis concurrent with persevering clinical decline strengthens our hypothesis that neuroinflammation importantly contributes to the Mfp2-/- phenotype.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis , Gliosis/etiology , Peroxisomal Multifunctional Protein-2/deficiency , Acoustic Stimulation , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/metabolism , Avoidance Learning/drug effects , Avoidance Learning/physiology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Encephalitis/complications , Encephalitis/genetics , Encephalitis/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Muscle Strength/drug effects , Muscle Strength/genetics , Peroxisomal Multifunctional Protein-2/genetics , Severity of Illness Index
17.
J Neuroinflammation ; 13(1): 230, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27590010

ABSTRACT

BACKGROUND: Although electroconvulsive therapy (ECT) is regarded as one of the efficient treatments for intractable psychiatric disorders, the mechanism of therapeutic action remains unclear. Recently, many studies indicate that ECT affects the immune-related cells, such as microglia, astrocytes, and lymphocytes. Moreover, microglial activation and astrocytic activation have been implicated in the postmortem brains of schizophrenia patients. We previously demonstrated that Gunn rats showed schizophrenia-like behavior and microglial activation in their brains. The present study examined the effects of electroconvulsive shock (ECS), an animal counterpart of ECT, on schizophrenia-like behavior, microgliosis, and astrogliosis in the brain of Gunn rats. METHODS: The rats were divided into four groups, i.e., Wistar sham, Wistar ECS, Gunn sham, and Gunn ECS. ECS groups received ECS once daily for six consecutive days. Subsequently, prepulse inhibition (PPI) test was performed, and immunohistochemistry analysis was carried out to determine the activation degree of microglia and astrocytes in the hippocampus by using anti-CD11b and anti-glial fibrillary acidic protein (GFAP) antibody, respectively. RESULTS: We found PPI deficit in Gunn rats compared to Wistar rats, and it was significantly improved by ECS. Immunohistochemistry analysis revealed that immunoreactivity of CD11b and GFAP was significantly increased in Gunn rats compared to Wistar rats. ECS significantly attenuated the immunoreactivity of both CD11b and GFAP in Gunn rats. CONCLUSIONS: ECS ameliorated schizophrenia-like behavior of Gunn rats and attenuated microgliosis and astrogliosis in the hippocampus of Gunn rats. Accordingly, therapeutic effects of ECT may be exerted, at least in part, by inhibition of glial activation. These results may provide crucial information to elucidate the role of activated glia in the pathogenesis of schizophrenia and to determine whether future therapeutic interventions should attempt to up-regulate or down-regulate glial functions.


Subject(s)
Electroshock , Gliosis/therapy , Hippocampus/pathology , Schizophrenia/pathology , Acoustic Stimulation , Animals , Astrocytes/pathology , Astrocytes/physiology , CD11b Antigen/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Hearing Disorders/genetics , Male , Microglia/pathology , Prepulse Inhibition/physiology , Psychoacoustics , Rats , Rats, Gunn , Rats, Wistar , Schizophrenia/complications , Schizophrenia/genetics
18.
Exp Brain Res ; 234(11): 3225-3232, 2016 11.
Article in English | MEDLINE | ID: mdl-27377070

ABSTRACT

We have reported previously that intracranial application of near-infrared light (NIr) reduces clinical signs and offers neuroprotection in a subacute MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) monkey model of Parkinson's disease. In this study, we explored whether NIr reduces the gliosis in this animal model. Sections of midbrain (containing the substantia nigra pars compacta; SNc) and striatum were processed for glial fibrillary acidic protein (to label astrocytes; GFAP) and ionised calcium-binding adaptor molecule 1 (to label microglia; IBA1) immunohistochemistry. Cell counts were undertaken using stereology, and cell body sizes were measured using ImageJ. Our results showed that NIr treatment reduced dramatically (~75 %) MPTP-induced astrogliosis in both the SNc and striatum. Among microglia, however, NIr had a more limited impact in both nuclei; although there was a reduction in overall cell size, there were no changes in the number of microglia in the MPTP-treated monkeys after NIr treatment. In summary, we showed that NIr treatment influenced the glial response, particularly that of the astrocytes, in our monkey MPTP model of Parkinson's disease. Our findings raise the possibility of glial cells as a future therapeutic target using NIr.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Gliosis/etiology , Gliosis/therapy , Infrared Rays/therapeutic use , MPTP Poisoning/complications , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Calcium-Binding Proteins , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Low-Level Light Therapy , MPTP Poisoning/pathology , Macaca fascicularis , Male , Microfilament Proteins , Neuroglia/drug effects , Neuroglia/radiation effects , Neurotoxins/toxicity , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
19.
Curr Opin Endocrinol Diabetes Obes ; 22(5): 325-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26192704

ABSTRACT

PURPOSE OF REVIEW: Hypothalamic inflammation and gliosis are recently discovered mechanisms that may contribute to obesity pathogenesis. Current research in this area suggests that investigation of these central nervous system responses may provide opportunities to develop new weight loss treatments. RECENT FINDINGS: In rodents, hypothalamic inflammation and gliosis occur rapidly with high-fat diet consumption prior to significant weight gain. In addition, sensitivity or resistance to diet-induced obesity in rodents generally correlates with the presence or absence of hypothalamic inflammation and reactive gliosis (brain response to injury). Moreover, functional interventions that increase or decrease inflammation in neurons and glia correspondingly alter diet-associated weight gain. However, some conflicting data have recently emerged that question the contribution of hypothalamic inflammation to obesity pathogenesis. Nevertheless, several studies have detected gliosis and disrupted connectivity in obese humans, highlighting the potential translational importance of this mechanism. SUMMARY: There is growing evidence that obesity is associated with brain inflammation in humans, particularly in the hypothalamus where its presence may disrupt body weight control and glucose homeostasis. More work is needed to determine whether this response is common in human obesity and to what extent it can be manipulated for therapeutic benefit.


Subject(s)
Encephalitis/pathology , Gliosis/pathology , Hypothalamus/pathology , Obesity/pathology , Animals , Diet, High-Fat/adverse effects , Encephalitis/etiology , Gliosis/etiology , Glucose/metabolism , Humans
20.
Cell Rep ; 9(6): 2124-38, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25497089

ABSTRACT

Diets rich in saturated fat produce inflammation, gliosis, and neuronal stress in the mediobasal hypothalamus (MBH). Here, we show that microglia mediate this process and its functional impact. Although microglia and astrocytes accumulate in the MBH of mice fed a diet rich in saturated fatty acids (SFAs), only the microglia undergo inflammatory activation, along with a buildup of hypothalamic SFAs. Enteric gavage specifically with SFAs reproduces microglial activation and neuronal stress in the MBH, and SFA treatment activates murine microglia, but not astrocytes, in culture. Moreover, depleting microglia abrogates SFA-induced inflammation in hypothalamic slices. Remarkably, depleting microglia from the MBH of mice abolishes inflammation and neuronal stress induced by excess SFA consumption, and in this context, microglial depletion enhances leptin signaling and reduces food intake. We thus show that microglia sense SFAs and orchestrate an inflammatory process in the MBH that alters neuronal function when SFA consumption is high.


Subject(s)
Astrocytes/metabolism , Dietary Fats/metabolism , Fatty Acids/metabolism , Hypothalamus/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Dietary Fats/adverse effects , Eating , Energy Metabolism , Fatty Acids/adverse effects , Gliosis/etiology , Gliosis/metabolism , Hypothalamus/cytology , Inflammation/etiology , Inflammation/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Neurons/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL