Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Sci Rep ; 12(1): 9389, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672451

ABSTRACT

Gut dysbiosis is closely associated with the outbreak of inflammatory bowel disease (IBD) and psychiatric disorder. The Enterobacteriaceae population was higher in the feces of patients with inflammatory bowel disease (IBD-F) than in those of healthy control volunteers (HC-F). The Enterococcaceae and Lactobacillaceae populations were higher in the feces of IBD patients with depression (IBD/D+-F) vs. the feces of IBD patients without depression (IBD/D--F). Therefore, we examined the effects of Klebsiella oxytoca, Escherichia coli, Cronobacter sakazakii, Enterococcus faecium, and Pediococcus acidolactici overpopulated in IBD/D+-F and their byproducts LPS and exopolysaccharide (EPS) on the occurrence of depression and colitis in mice. Oral gavages of Klebsiella oxytoca, Escherichia coli, and Cronobacter sakazakii belonging to Enterobacteriaceae, singly or together, caused dose-dependently colitis and depression-like behaviors in germ-free and specific-pathogen-free mice. Although Enterococcus faecium and Pediococcus acidolactici did not significantly cause colitis and depression-like behaviors, they significantly deteriorated Klebsiella oxytoca- or Escherichia coli-induced colitis, neuroinflammation, and anxiety/depression-like behaviors and increased blood LPS, corticosterone, and IL-6 levels. The EPSs from Enterococcus faecium and Pediococcus acidolactici also worsened Klebsiella oxytoca LPS-induced colitis, neuroinflammation, and depression-like behaviors in mice and increased the translocation of fluorescein isothiocyanate-conjugated LPS into the hippocampus. However, Bifidobacterium longum, which was lower in IBD/D+-F vs. IBD/D--F, or its EPS suppressed them. In conclusion, Enterococcus faecium and Pediococcus acidolactici, known as a probiotic strain, and their EPSs may be a risk factor for the outbreak of depression and IBD.


Subject(s)
Colitis , Enterococcus faecium , Inflammatory Bowel Diseases , Pediococcus acidilactici , Animals , Colitis/chemically induced , Colitis/microbiology , Depression/psychology , Enterobacteriaceae , Escherichia coli , Humans , Lipopolysaccharides , Mice
4.
Front Microbiol ; 11: 564271, 2020.
Article in English | MEDLINE | ID: mdl-33324357

ABSTRACT

Gut microbiota is closely associated with the occurrence of neuropsychiatric disorders. Antibiotics are frequently used to prevent pathogen infection in patients with brain ischemia. To understand the impact of prophylactic antibiotic treatment for patients with brain ischemia, we examined the effects of orally administered vancomycin and ampicillin on cognitive function and gut microbiota composition in mice with transient global forebrain ischemia (tIsc). tIsc operation and orally gavaged vancomycin mildly and moderately caused cognitive impairment, respectively. However, the exposure of mice with tIsc to vancomycin or ampicillin severely impaired cognitive function in the Y-maze, novel object recognition, and Banes maze tasks. Furthermore, their treatments induced NF-κB activation as well as active microglia (NF-κB+/Iba1+ and LPS+/Iba1+ cells) and apoptotic (caspase 3+/NeuN+) cell population in the hippocampus, whereas the brain-derived neurotrophic factor (BDNF)+/NeuN+ cell populations decreased. These treatments also caused colitis and gut dysbiosis. They increased the population of Proteobacteria including Enterobacter xiangfangenesis. Orally delivered fecal transplantation of vancomycin-treated mice with or without tIsc and oral gavage of Enterobacter xiangfangenesis also significantly deteriorated the cognitive impairment and colitis in transplanted mice with tIsc. These findings suggest that oral administration of antibiotics can deteriorate cognitive impairment with gut dysbiosis in patients with brain ischemia.

5.
Nutrients ; 12(11)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182607

ABSTRACT

Excessive expression of interleukin (IL)-1ß in the brain causes depression and cognitive dysfunction. Herein, we investigated the effect of Lactobacillus gasseri NK109, which suppressed IL-1ß expression in activated macrophages, on Escherichia coli K1-induced cognitive impairment and depression in mice. Germ-free and specific pathogen-free mice with neuropsychiatric disorders were prepared by oral gavage of K1. NK109 alleviated K1-induced cognition-impaired and depressive behaviors, decreased the expression of IL-1ß and populations of NF-κB+/Iba1+ and IL-1R+ cells, and increased the K1-suppressed population of BDNF+/NeuN+ cells in the hippocampus. However, its effects were partially attenuated by celiac vagotomy. NK109 treatment mitigated K1-induced colitis and gut dysbiosis. Tyndallized NK109, even if lysed, alleviated cognitive impairment and depression. In conclusion, NK109 alleviated neuropsychiatric disorders and colitis by modulating IL-1ß expression, gut microbiota, and vagus nerve-mediated gut-brain signaling.


Subject(s)
Colitis/therapy , Escherichia coli/physiology , Gastrointestinal Microbiome/physiology , Interleukin-1beta/metabolism , Lactobacillus gasseri/physiology , Microbial Interactions/physiology , Animals , Behavior, Animal , Cognitive Dysfunction , Colitis/psychology , Depression , Dysbiosis/psychology , Dysbiosis/therapy , Escherichia coli/immunology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Hippocampus/drug effects , Hippocampus/metabolism , Lactobacillus gasseri/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Probiotics/administration & dosage
6.
Microbiome ; 8(1): 107, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669127

ABSTRACT

BACKGROUND: In a pilot study, we found that feces transplantation from elderly individuals to mice significantly caused cognitive impairment. Paenalcaligenes hominis and Escherichia coli are increasingly detected in the feces of elderly adults and aged mice. Therefore, we isolated Paenalcaligenes hominis and Escherichia coli from the feces of elderly individuals and aged mice and examined their effects on the occurrence of age-related degenerative cognitive impairment and colonic inflammation in mice. RESULTS: The transplantation of feces collected from elderly people and aged mice caused significantly more severe cognitive impairment in transplanted young mice than those from young adults and mice. Oral gavage of Paenalcaligenes hominis caused strong cognitive impairment and colitis in specific pathogen-free (SPF) and germ-free mice. Escherichia coli also induced cognitive impairment and colitis in SPF mice. Oral gavage of Paenalcaligenes hominis, its extracellular vesicles (EVs), and/or lipopolysaccharide caused cognitive impairment and colitis in mice. However, celiac vagotomy significantly inhibited the occurrence of cognitive impairment, but not colitis, in mice exposed to Paenalcaligenes hominis or its EVs, whereas its lipopolysaccharide or Escherichia coli had no such effects. Vagotomy also inhibited the infiltration of EVs into the hippocampus. CONCLUSIONS: Paenalcaligenes hominis, particularly its EVs, can cause cognitive function-impaired disorders, such as Alzheimer's disease, and its EVs may penetrate the brain through the blood as well as the vagus nerve. Video Abstract.


Subject(s)
Alcaligenaceae/pathogenicity , Cognitive Dysfunction/etiology , Cognitive Dysfunction/microbiology , Extracellular Vesicles , Intestines/microbiology , Vagus Nerve , Aging/metabolism , Animals , Colitis/etiology , Colitis/microbiology , Escherichia coli/pathogenicity , Fecal Microbiota Transplantation , Feces/microbiology , Female , Gastrointestinal Microbiome , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pilot Projects , Risk Factors , Vagotomy , Young Adult
7.
J Microbiol Biotechnol ; 30(8): 1222-1226, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32347078

ABSTRACT

Lactobacillus reuteri NK33 (NK33) and Bifidobacterium adolescentis NK98 (NK98) alleviate immobilization stress-induced depression. To understand the gut microbiota-mediated mechanisms of NK33 and NK98 against depression, we examined their effects on Escherichia coli K1 (K1)-induced depression and gut dysbiosis in mice. NK33, NK98, and their mixtures (1:1, 4:1, and 9:1) mitigated K1-induced depression and colitis. NK33 and NK98 additively or synergistically increased BDNF+/NeuN+ cell population and suppressed NF-κB action in the hippocampus. They alleviated gut dysbiosis by reducing the Proteobacteria population and increasing the Clostridia population. These results suggest that NK33 and NK98 may alleviate depression and colitis by ameliorating gut dysbiosis.


Subject(s)
Bifidobacterium adolescentis/physiology , Depression/therapy , Dysbiosis/therapy , Escherichia coli/pathogenicity , Gastrointestinal Microbiome/physiology , Limosilactobacillus reuteri/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Colitis/microbiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Depression/microbiology , Disease Models, Animal , Dysbiosis/microbiology , Feces/microbiology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
8.
Front Immunol ; 11: 273, 2020.
Article in English | MEDLINE | ID: mdl-32158447

ABSTRACT

To understand the roles of human gut bacteria in the occurrence of neuropsychiatric disorders, we isolated inflammatory Escherichia coli K1 and anti-inflammatory Lactobacillus mucosae from healthy human feces and examined their effects on the occurrence of altered microbiota, cognitive decline, and depression in mice. Oral gavage of Escherichia coli K1 caused colitis, cognitive decline, and depression in mice in the elevated plus maze, tail suspension, and forced swimming tasks. However, NK41 treatment reduced K1-induced cognitive decline and anxiety/depression. Furthermore, NK41 treatment increased K1-suppressed brain-derived neurotrophic factor (BDNF) expression and BDNF+/NeuN+ cell population and suppressed K1-induced NF-κB activation and LPS+/Iba1+ and NF-κB+/Iba1+ (microglial) cell populations in the hippocampus. NK41 treatment also suppressed K1-induced TNF-α and LPS levels in the blood and TNF-α expression, myeloperoxidase activity, NF-κB+/CD11c+ and CD11b+/CD11c+ cell populations in the colon. Furthermore, NK41 treatment decreased K1-induced colonic MUC2 expression, gut Proteobacteria population, and fecal LPS levels and modified the bacterial abundance related to polysaccharide breaking and biosynthesis. In conclusion, the overgrowth of inflammatory bacteria such as Escherichia coli in the gastrointestinal tract can cause neuropsychiatric disorders with gut microbiota alteration and the superiority of anti-inflammatory bacteria such as Lactobacillus mucosae can alleviate neuropsychiatric disorders with the attenuation of altered microbiota.


Subject(s)
Escherichia coli/physiology , Gastrointestinal Microbiome/physiology , Lactobacillus/physiology , Microbial Interactions/physiology , Animals , Anxiety , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction , Depression , Escherichia coli/immunology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation , Lactobacillus/immunology , Macrophages , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Exp Neurobiol ; 29(1): 70-79, 2020 Feb 29.
Article in English | MEDLINE | ID: mdl-32122109

ABSTRACT

Many previous studies have shown reduced glucose uptake in the ischemic brain. In contrast, in a permanent unilateral common carotid artery occlusion (UCCAO) mouse model, our pilot experiments using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) revealed that a subset of mice exhibited conspicuously high uptake of glucose in the ipsilateral hemisphere at 1 week post-occlusion (asymmetric group), whereas other mice showed symmetric uptake in both hemispheres (symmetric group). Thus, we aimed to understand the discrepancy between the two groups. Cerebral blood flow and histological/metabolic changes were analyzed using laser Doppler flowmetry and immunohistochemistry/Western blotting, respectively. Contrary to the increased glucose uptake observed in the ischemic cerebral hemisphere on FDG PET (p<0.001), cerebral blood flow tended to be lower in the asymmetric group than in the symmetric group (right to left ratio [%], 36.4±21.8 vs. 58.0±24.8, p=0.059). Neuronal death was observed only in the ischemic hemisphere of the asymmetric group. In contrast, astrocytes were more activated in the asymmetric group than in the symmetric group (p<0.05). Glucose transporter-1, and monocarboxylate transporter-1 were also upregulated in the asymmetric group, compared with the symmetric group (p<0.05, respectively). These results suggest that the increased FDG uptake was associated with relatively severe ischemia, and glucose transporter-1 upregulation and astrocyte activation. Glucose metabolism may thus be a compensatory mechanism in the moderately severe ischemic brain.

10.
Sci Rep ; 9(1): 11814, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413350

ABSTRACT

To understand the role of commensal gut bacteria on the progression of cognitive decline in Alzheimer's disease via the microbiota-gut-brain axis, we isolated anti-inflammatory Bifidobacterium longum (NK46) from human gut microbiota, which potently inhibited gut microbiota endotoxin production and suppressed NF-κB activation in lipopolysaccharide (LPS)-stimulated BV-2 cells, and examined whether NK46 could simultaneously alleviate gut dysbiosis and cognitive decline in male 5xFAD-transgenic (5XFAD-Tg, 6 months-old) and aged (18 months-old) mice. Oral administration of NK46 (1 × 109 CFU/mouse/day for 1 and 2 months in aged and Tg mice, respectively) shifted gut microbiota composition, particularly Proteobacteria, reduced fecal and blood LPS levels, suppressed NF-κB activation and TNF-α expression, and increased tight junction protein expression in the colon of 5XFAD-Tg and aged mice. NK46 treatment also alleviated cognitive decline in 5XFAD-Tg and aged mice. Furthermore, NK46 treatment suppressed amyloid-ß, ß/γ-secretases, and caspase-3 expression and amyloid-ß accumulation in the hippocampus of 5XFAD-Tg mice. NK46 treatment also reduced Iba1+, LPS+/CD11b+, and caspase-3+/NeuN+ cell populations and suppressed NF-κB activation in the hippocampus of 5XFAD-Tg and aged mice, while BDNF expression was increased. These findings suggest that the suppression of gut dysbiosis and LPS production by NK46 can mitigate cognitive decline through the regulation of microbiota LPS-mediated NF-κB activation.


Subject(s)
Aging/physiology , Bifidobacterium longum/physiology , Cognitive Dysfunction/prevention & control , Dysbiosis , Gastrointestinal Microbiome , Probiotics/administration & dosage , Animals , Cognitive Dysfunction/microbiology , Feces/microbiology , Humans , Lipopolysaccharides/metabolism , Mice , Mice, Transgenic
11.
Neurobiol Dis ; 132: 104538, 2019 12.
Article in English | MEDLINE | ID: mdl-31344491

ABSTRACT

It is challenging to revitalize ischemic penumbra after an acute stroke with intracranial perfusion insufficiency. To evaluate whether cranial burr hole and erythropoietin (EPO) generate effective revascularization, we investigated the efficacy of the augmentation method for reverse arteriogenesis from the healthy extracranial milieu. An intracranial perfusion insufficiency was created through bilateral internal carotid artery ligation (bICAL) in Sprague-Dawley rats. We administered recombinant human EPO (5000 U/kg) or saline intraperitoneally for 3 days after bICAL. Mechanical barrier disruption (MBD) was performed through a cranial burr hole with small dural cracks in the right hemisphere. The ipsilateral hemisphere with MBD grossly showed vascular networks between the extra- and intra-cranial spaces 2 weeks after the MBD procedure. It also showed significantly increased vessels in the intracranial vasculature adjacent to the MBD region (p = 0.0006). The levels of pro-angiogenic and inflammatory factors with prominent markers of vessel permeability were also significantly increased (MBD-only vs. control; Tnf-α, p = 0.0007; Vegf, p = 0.0206). In the EPO-administered group, such elevations in inflammation were significantly mitigated (combined vs. MBD-only; Tnf-α, p = 0.0008). The ipsilateral hemisphere with MBD-EPO (vs. MBD-only) showed significantly increased vessels (RECA-1, p = 0.0182) and their maturation (RECA-1/α-SMA, p = 0.0046), with upregulation of tumor growth factor-ß1 (Tgf-ß1, p = 0.037) and matrix metalloproteinase-2 (Mmp-2, p = 0.0488). These findings were completely blocked by minocycline (MIC) administration during in vivo (Tgf-ß1, p = 0.0009; Mmp-2, p < 0.0001) and in vitro experiments (tube formation, p < 0.0001). Our data suggest that the MBD procedure (for angiogenic routes) and EPO administration (for an arteriogenic booster) are complimentary and can facilitate successfully "reverse arteriogenesis" in subjects with intracranial perfusion insufficiency.


Subject(s)
Cerebral Arteries/diagnostic imaging , Cerebral Revascularization/methods , Craniotomy/methods , Erythropoietin/administration & dosage , Moyamoya Disease/diagnostic imaging , Moyamoya Disease/drug therapy , Adult , Aged , Animals , Cerebral Angiography/methods , Cerebral Arteries/drug effects , Combined Modality Therapy/methods , Human Umbilical Vein Endothelial Cells , Humans , Male , Middle Aged , Prospective Studies , Random Allocation , Rats , Rats, Sprague-Dawley , Retrospective Studies , Skull/blood supply , Skull/diagnostic imaging , Skull/drug effects
12.
Nutrients ; 11(4)2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30979031

ABSTRACT

The gut dysbiosis by stressors such as immobilization deteriorates psychiatric disorders through microbiota-gut-brain axis activation. To understand whether probiotics could simultaneously alleviate anxiety/depression and colitis, we examined their effects on immobilization stress (IS)-induced anxiety/depression and colitis in mice. The probiotics Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 were isolated from healthy human feces. Mice with anxiety/depression and colitis were prepared by IS treatment. NK33 and NK98 potently suppressed NF-κB activation in lipopolysaccharide (LPS)-induced BV-2 cells. Treatment with NK33 and/or NK98, which were orally gavaged in mice before or after IS treatment, significantly suppressed the occurrence and development of anxiety/depression, infiltration of Iba1⁺ and LPS⁺/CD11b⁺ cells (activated microglia) into the hippocampus, and corticosterone, IL-6, and LPS levels in the blood. Furthermore, they induced hippocampal BDNF expression while NF-κB activation was suppressed. NK33 and/or NK98 treatments suppressed IS-induced colon shortening, myeloperoxidase activity, infiltration of CD11b⁺/CD11c⁺ cells, and IL-6 expression in the colon. Their treatments also suppressed the IS-induced fecal Proteobacteria population and excessive LPS production. They also induced BDNF expression in LPS-induced SH-SY5Y cells in vitro. In conclusion, NK33 and NK98 synergistically alleviated the occurrence and development of anxiety/depression and colitis through the regulation of gut immune responses and microbiota composition.


Subject(s)
Anxiety/therapy , Bifidobacterium adolescentis , Colitis/therapy , Depression/therapy , Limosilactobacillus reuteri , Stress, Psychological/complications , Animals , Anxiety/prevention & control , Anxiety/psychology , Cell Line , Colitis/psychology , Depression/prevention & control , Depression/psychology , Dysbiosis/psychology , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia , NF-kappa B/metabolism , Neuroblastoma , Probiotics/administration & dosage , Restraint, Physical , Stress, Psychological/etiology , Tumor Cells, Cultured
13.
Sci Rep ; 8(1): 13897, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224732

ABSTRACT

The present study aimed to understand the crosstalk between anxiety and gut microbiota. Exposure of mice to immobilization stress (IS) led to anxiety-like behaviors, increased corticosterone and tumor necrosis factor-α levels in the blood, increased nuclear factor (NF)-κB activation and microglia/monocyte populations in the hippocampus, and suppressed brain-derived neurotrophic factor (BDNF) expression in the hippocampus. Furthermore, IS exposure increased NF-κB activation and monocyte population in the colon and increased Proteobacteria and Escherichia coli populations in the gut microbiota and fecal and blood lipopolysaccharide (LPS) levels while decreasing the lactobacilli population. Oral administration of the fecal microbiota of mice treated with IS (FIS) or E. coli led to the increased NF-κB activation and monocyte population in the colon. These treatments increased blood corticosterone and LPS levels and anxiety-like behaviors, decreased BDNF expression, and induced NF-κB activation and microglia/monocyte populations in the hippocampus. Intraperitoneal injection of LPS purified from E. coli also led to anxiety and colitis in mice. Oral administration of commensal lactobacilli, particularly Lactobacillus johnsonii, attenuated IS- or E. coli-induced colitis and anxiety-like behaviors and biomarkers. These findings suggest that exposure to stressors can increase Proteobacteria populations and fecal LPS levels and cause gastrointestinal inflammation, resulting in the deterioration of anxiety through NF-κB activation. However, the amelioration of gastrointestinal inflammation by treatment with probiotics including L. johnsonii can alleviate anxiety.


Subject(s)
Anxiety/etiology , Escherichia coli/growth & development , Gastrointestinal Microbiome , Immobilization/psychology , NF-kappa B/metabolism , Stress, Physiological , Animals , Anxiety/microbiology , Colitis/etiology , Colitis/microbiology , Colitis/therapy , Fecal Microbiota Transplantation , Male , Mice , Mice, Inbred C57BL , Probiotics/administration & dosage
14.
Korean J Physiol Pharmacol ; 20(2): 185-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26937215

ABSTRACT

Ampicillin, a ß-lactam antibiotic, dose-dependently protects neurons against ischemic brain injury. The present study was performed to investigate the neuroprotective mechanism of ampicillin in a mouse model of transient global forebrain ischemia. Male C57BL/6 mice were anesthetized with halothane and subjected to bilateral common carotid artery occlusion for 40 min. Before transient forebrain ischemia, ampicillin (200 mg/kg, intraperitoneally [i.p.]) or penicillin G (6,000 U/kg or 20,000 U/kg, i.p.) was administered daily for 5 days. The pretreatment with ampicillin but not with penicillin G signifi cantly attenuated neuronal damage in the hippocampal CA1 subfield. Mechanistically, the increased activity of matrix metalloproteinases (MMPs) following forebrain ischemia was also attenuated by ampicillin treatment. In addition, the ampicillin treatment reversed increased immunoreactivities to glial fibrillary acidic protein and isolectin B4, markers of astrocytes and microglia, respectively. Furthermore, the ampicillin treatment significantly increased the level of glutamate transporter-1, and dihydrokainic acid (DHK, 10 mg/kg, i.p.), an inhibitor of glutamate transporter-1 (GLT-1), reversed the neuroprotective effect of ampicillin. Taken together, these data indicate that ampicillin provides neuroprotection against ischemia-reperfusion brain injury, possibly through inducing the GLT-1 protein and inhibiting the activity of MMP in the mouse hippocampus.

15.
Glia ; 60(12): 1915-29, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22907804

ABSTRACT

The Bcl-2-interacting death suppressor (Bis) protein is involved in antiapoptosis and antistress pathways. However, its roles after neonatal hypoxia-ischemia remain obscure. Therefore, we investigated the effects of Bis deletion on hippocampal cell death following neonatal hypoxia-ischemia. We transected the right common carotid artery of bis(+/+) and bis(-/-) mice at postnatal Day 7 and subjected them to hypoxia for 35 min. Cresyl violet staining showed that hypoxia-ischemia induced progressive cell death in the hippocampi of bis(+/+) mice. Moreover, Bis was expressed in astrocytes, not microglia, in sham-manipulated hippocampi of bis(+/+) mice, and was markedly enhanced after hypoxia-ischemia. Immunoblotting showed that Bis expression significantly increased 3 and 7 days following hypoxia-ischemia. Unexpectedly, 7 days after hypoxia-ischemia, the number of hippocampal NeuN-positive cells was higher in the bis(-/-) mice than in the bis(+/+) mice. We subsequently performed transcriptomic analysis and quantitative real time polymerase chain reaction to search for the underlying genes responsible for resistance to hypoxia-ischemia in the bis(-/-) hippocampus. These studies showed that 6 h after hypoxia-ischemia, galectin 3 and filamin C levels increased to a lesser extent in the bis(-/-) hippocampi compared with the bis(+/+) hippocampi. Finally, our in vitro hypoxia-ischemia model, using A172 glioma cells and primary astrocytes, showed that downregulation of Bis blocked the enhanced expression of galectin 3 after oxygen-glucose deprivation. This study demonstrated that Bis was upregulated in the astrocytes after hypoxia-ischemia. In addition, we showed that hippocampal neurons are less vulnerable to hypoxia-ischemia in mice lacking Bis, possibly because of the modulation of galectin 3 induction.


Subject(s)
Carrier Proteins/genetics , Down-Regulation/genetics , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Apoptosis Regulatory Proteins , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Cell Line, Tumor , Hippocampus/pathology , Humans , Hypoxia-Ischemia, Brain/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Rats , Rats, Sprague-Dawley
16.
Korean J Physiol Pharmacol ; 12(6): 287-91, 2008 12.
Article in English | MEDLINE | ID: mdl-19967069

ABSTRACT

Ampicillin, a beta-lactam antibiotic, has been reported to induce astrocytic glutamate transporter-1 which plays a crucial role in protecting neurons against glutamate excitotoxicity. We investigated the effect of ampicillin on neuronal damage in the mouse hippocampus and neostriatum following transient global forebrain ischemia. Male C57BL/6 mice were anesthetized with halothane and subjected to bilateral occlusion of the common carotid artery for 40 min. Ampicillin was administered post-ischemically (for 3 days) and/or pre-ischemically (for 3~5 days until one day before the onset of ischemia). Pre- and post-ischemic treatment with ampicillin (50 mg/kg/day or 200 mg/kg/day) prevented ischemic neuronal death in the medial CA1 area of the hippocampus as well as the neostriatum in a dose-dependent manner. In addition, ischemic neuronal damage was reduced by pre-ischemic treatment with ampicillin (200 mg/kg/day). In summary, our results suggest that ampicillin plays a functional role as a chemical preconditioning agent that protects hippocampal neurons from ischemic insult.

SELECTION OF CITATIONS
SEARCH DETAIL
...