Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Nat Metab ; 2(8): 688-702, 2020 08.
Article in English | MEDLINE | ID: mdl-32694825

ABSTRACT

Adipose tissue eosinophils (ATEs) are important in the control of obesity-associated inflammation and metabolic disease. However, the way in which ageing impacts the regulatory role of ATEs remains unknown. Here, we show that ATEs undergo major age-related changes in distribution and function associated with impaired adipose tissue homeostasis and systemic low-grade inflammation in both humans and mice. We find that exposure to a young systemic environment partially restores ATE distribution in aged parabionts and reduces adipose tissue inflammation. Approaches to restore ATE distribution using adoptive transfer of eosinophils from young mice into aged recipients proved sufficient to dampen age-related local and systemic low-grade inflammation. Importantly, restoration of a youthful systemic milieu by means of eosinophil transfers resulted in systemic rejuvenation of the aged host, manifesting in improved physical and immune fitness that was partially mediated by eosinophil-derived IL-4. Together, these findings support a critical function of adipose tissue as a source of pro-ageing factors and uncover a new role of eosinophils in promoting healthy ageing by sustaining adipose tissue homeostasis.


Subject(s)
Adipose Tissue/physiology , Eosinophils/physiology , Immunity , Inflammation/pathology , Physical Fitness/physiology , Adipose Tissue/pathology , Adipose Tissue, White/pathology , Adipose Tissue, White/physiology , Adult , Aged , Aging , Animals , Eosinophils/immunology , Eosinophils/pathology , Gene Expression Regulation , Glucose Tolerance Test , Homeostasis , Humans , Interleukin-4/immunology , Interleukin-4/physiology , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Strength , Satellite Cells, Skeletal Muscle/metabolism , Young Adult
3.
J Colloid Interface Sci ; 528: 18-26, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-29807352

ABSTRACT

Developing efficient bi-functional electrocatalysts for both oxygen reduction reaction (ORR) and hydrogen evolution reaction (HER) is crucial for producing hydrogen and utilizing hydrogen effectively to promote electrochemical energy storage in proton membrane exchange fuel cells (PEMFCs). Herein, we report Co@Pd core-shell nanoparticles encapsulated in porous carbon derived from zeolitic imidazolate framework 67 (ZIF-67) for both ORR and HER. The controlled pyrolysis of ZIF-67 can lead to the formation of Co nanoparticles encapsulated in nitrogen-doped porous carbon (Co NC), which subsequently underwent galvanic replacement with Na2PdCl4 to form Co@Pd core-shell nanoparticles embedded in nitrogen-doped porous carbon (Co@Pd NC). The Co@Pd NC exhibited outperformance in ORR and HER than commercial Pd/C, as manifested by more positive onset potential and larger diffusion-limited current density in ORR tests, as well as a small overpotential to drive a current density of 10 mA cm-2, and much lower Tafel slope in HER tests. It also demonstrated more robust long-term stability than commercial Pd/C for both ORR and HER. Multiple techniques inter-confirmed that the Pd loading in the sample was very low. The findings can pave a path for fabricating a core-shell structured nanocomposite with ultralow noble metal usage as a bifunctional catalyst for electrochemical energy storage and conversion with high-efficiency and remarkable longevity.

4.
J Neuroinflammation ; 15(1): 55, 2018 Feb 22.
Article in English | MEDLINE | ID: mdl-29471880

ABSTRACT

BACKGROUND: The cystine/glutamate antiporter (xc-) has been implicated in several neurological disorders and, specifically, in multiple sclerosis (MS) as a mediator of glutamate excitotoxicity and proinflammatory immune responses. We aimed to evaluate an xc-specific positron emission tomography (PET) radiotracer, (4S)-4-(3-[18F]fluoropropyl)-L-glutamate ([18F]FSPG), for its ability to allow non-invasive monitoring of xc- activity in a mouse model of MS. METHODS: Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice by subcutaneous injection of myelin oligodendrocyte glycoprotein (MOG35-55) peptide in complete Freund's adjuvant (CFA) followed by pertussis toxin. Control mice received CFA emulsion and pertussis toxin without MOG peptide, while a separate cohort of naïve mice received no treatment. PET studies were performed to investigate the kinetics and distribution of [18F]FSPG in naïve, control, pre-symptomatic, and symptomatic EAE mice, compared to 18F-fluorodeoxyglucose ([18F]FDG). After final PET scans, each mouse was perfused and radioactivity in dissected tissues was measured using a gamma counter. Central nervous system (CNS) tissues were further analyzed using ex vivo autoradiography or western blot. [18F]FSPG uptake in human monocytes, and T cells pre- and post-activation was investigated in vitro. RESULTS: [18F]FSPG was found to be more sensitive than [18F]FDG at detecting pathological changes in the spinal cord and brain of EAE mice. Even before clinical signs of disease, a small but significant increase in [18F]FSPG signal was observed in the spinal cord of EAE mice compared to controls. This increase in PET signal became more pronounced in symptomatic EAE mice and was confirmed by ex vivo biodistribution and autoradiography. Likewise, in the brain of symptomatic EAE mice, [18F]FSPG uptake was significantly higher than controls, with the largest changes observed in the cerebellum. Western blot analyses of CNS tissues revealed a significant correlation between light chain of xc- (xCT) protein levels, the subunit of xc- credited with its transporter activity, and [18F]FSPG-PET signal. In vitro [18F]FSPG uptake studies suggest that both activated monocytes and T cells contribute to the observed in vivo PET signal. CONCLUSION: These data highlight the promise of [18F]FSPG-PET as a technique to provide insights into neuroimmune interactions in MS and the in vivo role of xc- in the development and progression of this disease, thus warranting further investigation.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fluorine Radioisotopes/metabolism , Glutamates/metabolism , Positron-Emission Tomography/methods , Animals , Cells, Cultured , Fluorodeoxyglucose F18/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/metabolism
5.
J Agric Food Chem ; 64(12): 2498-505, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26551795

ABSTRACT

Bamboo shoots are a delicacy in Asia. Two novel compounds, adenine-(1'R,2'R,3'R)-cyclic butanetetraol carbonate (16) and (-)-(7R,8S)-(4-hydroxy-3-methoxyphenylglycerol 9-O-ß-D-[6-O-4-hydroxy-3-methoxybenzoyl])-glucopyranoside (20), together with 12 known nucleosides (1-12), 3 amino acids (13-15), ß-carboline (17), and 2 megastigmane glycosides (18, 19) were isolated from bamboo shoots (Phyllostachys pubescens). Their structures and absolute configurations were rigorously determined by detailed spectroscopic analysis, and the composition of carbohydrates in bamboo shoots was qualitatively detected and quantitatively analyzed with ion chromatography. A simple, rapid, sensitive, and accurate HPLC-UV analysis was built for routine edible quality control of bamboo shoots, and 12 major components of bamboo shoots were quantitatively analyzed. The major chemical constituents of bamboo shoots were determined to be carbohydrates, amino acids, and nucleotides. These findings are correctives to the usual view of bamboo shoots chemical composition, and the previous research reports about the chemical composition of bamboo shoots may have taken the aromatic amino acids and nucleotides for flavonoids and phenolic acids.


Subject(s)
Drugs, Chinese Herbal/chemistry , Glycosides/isolation & purification , Poaceae/chemistry , Amino Acids/analysis , Carbohydrates/analysis , Carbolines/analysis , Chromatography, High Pressure Liquid , Cyclohexanones/analysis , Glucosides/analysis , Glycosides/chemistry , Molecular Structure , Norisoprenoids/analysis , Nuclear Magnetic Resonance, Biomolecular , Nucleotides/analysis , Stereoisomerism
6.
Science ; 349(6254): 1301-6, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26303961

ABSTRACT

The identification of human broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem revitalized hopes of developing a universal influenza vaccine. Using a rational design and library approach, we engineered stable HA stem antigens ("mini-HAs") based on an H1 subtype sequence. Our most advanced candidate exhibits structural and bnAb binding properties comparable to those of full-length HA, completely protects mice in lethal heterologous and heterosubtypic challenge models, and reduces fever after sublethal challenge in cynomolgus monkeys. Antibodies elicited by this mini-HA in mice and nonhuman primates bound a wide range of HAs, competed with human bnAbs for HA stem binding, neutralized H5N1 viruses, and mediated antibody-dependent effector activity. These results represent a proof of concept for the design of HA stem mimics that elicit bnAbs against influenza A group 1 viruses.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Humans , Mice , Protein Multimerization , Protein Structure, Secondary
7.
J Nucl Med ; 56(2): 311-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25613536

ABSTRACT

UNLABELLED: Herein we aimed to evaluate the utility of N-(2,5-dimethoxybenzyl)-2-(18)F-fluoro-N-(2-phenoxyphenyl)acetamide ((18)F-PBR06) for detecting alterations in translocator protein (TSPO) (18 kDa), a biomarker of microglial activation, in a mouse model of Alzheimer's disease (AD). METHODS: Wild-type (wt) and AD mice (i.e., APP(L/S)) underwent (18)F-PBR06 PET imaging at predetermined time points between the ages of 5-6 and 15-16 mo. MR images were fused with PET/CT data to quantify (18)F-PBR06 uptake in the hippocampus and cortex. Ex vivo autoradiography and TSPO/CD68 immunostaining were also performed using brain tissue from these mice. RESULTS: PET images showed significantly higher accumulation of (18)F-PBR06 in the cortex and hippocampus of 15- to 16-mo-old APP(L/S) mice than age-matched wts (cortex/muscle: 2.43 ± 0.19 vs. 1.55 ± 0.15, P < 0.005; hippocampus/muscle: 2.41 ± 0.13 vs. 1.55 ± 0.12, P < 0.005). And although no significant difference was found between wt and APP(L/S) mice aged 9-10 mo or less using PET (P = 0.64), we were able to visualize and quantify a significant difference in (18)F-PBR06 uptake in these mice using autoradiography (cortex/striatum: 1.13 ± 0.04 vs. 0.96 ± 0.01, P < 0.05; hippocampus/striatum: 1.266 ± 0.003 vs. 1.096 ± 0.017, P < 0.001). PET results for 15- to 16-mo-old mice correlated well with autoradiography and immunostaining (i.e., increased (18)F-PBR06 uptake in brain regions containing elevated CD68 and TSPO staining in APP(L/S) mice, compared with wts). CONCLUSION: (18)F-PBR06 shows great potential as a tool for visualizing TSPO/microglia in the progression and treatment of AD.


Subject(s)
Acetanilides , Alzheimer Disease/metabolism , Cerebral Cortex/diagnostic imaging , Hippocampus/diagnostic imaging , Receptors, GABA/metabolism , Alzheimer Disease/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Autoradiography , Biomarkers/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Disease Progression , Hippocampus/metabolism , Hippocampus/pathology , Magnetic Resonance Imaging , Mice , Muscles/metabolism , Positron-Emission Tomography , Reproducibility of Results , Time Factors
8.
Front Neurol ; 5: 12, 2014.
Article in English | MEDLINE | ID: mdl-24550885

ABSTRACT

Mild traumatic brain injury (mTBI, also referred to as concussion) accounts for the majority of all traumatic brain injuries. The consequences of repetitive mTBI have become of particular concern for individuals engaged in certain sports or in military operations. Many mTBI patients suffer long-lasting neurobehavioral impairments. In order to expedite pre-clinical research and therapy development, there is a need for animal models that reflect the long-term cognitive and pathological features seen in patients. In the present study, we developed and characterized a mouse model of repetitive mTBI, induced onto the closed head over the left frontal hemisphere with an electromagnetic stereotaxic impact device. Using GFAP-luciferase bioluminescence reporter mice that provide a readout of astrocyte activation, we observed an increase in bioluminescence relative to the force delivered by the impactor after single impact and cumulative effects of repetitive mTBI. Using the injury parameters established in the reporter mice, we induced a repetitive mTBI in wild-type C57BL/6J mice and characterized the long-term outcome. Animals received repetitive mTBI showed a significant impairment in spatial learning and memory when tested at 2 and 6 months after injury. A robust astrogliosis and increased p-Tau immunoreactivity were observed upon post-mortem pathological examinations. These findings are consistent with the deficits and pathology associated with mTBI in humans and support the use of this model to evaluate potential therapeutic approaches.

9.
J Exp Med ; 211(2): 189-98, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24493798

ABSTRACT

Aberrant microglial responses contribute to neuroinflammation in many neurodegenerative diseases, but no current therapies target pathogenic microglia. We discovered unexpectedly that the antiviral drug ganciclovir (GCV) inhibits the proliferation of microglia in experimental autoimmune encephalomyelitis (EAE), a mouse model for multiple sclerosis (MS), as well as in kainic acid-induced excitotoxicity. In EAE, GCV largely prevented infiltration of T lymphocytes into the central nervous system (CNS) and drastically reduced disease incidence and severity when delivered before the onset of disease. In contrast, GCV treatment had minimal effects on peripheral leukocyte distribution in EAE and did not inhibit generation of antibodies after immunization with ovalbumin. Additionally, a radiolabeled analogue of penciclovir, [(18)F]FHBG, which is similar in structure to GCV, was retained in areas of CNS inflammation in EAE, but not in naive control mice, consistent with the observed therapeutic effects. Our experiments suggest GCV may have beneficial effects in the CNS beyond its antiviral properties.


Subject(s)
Antiviral Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ganciclovir/pharmacology , Microglia/drug effects , Animals , Antiviral Agents/pharmacokinetics , Brain/drug effects , Brain/immunology , Brain/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Ganciclovir/pharmacokinetics , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Microglia/pathology , Ovalbumin/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
10.
J Exp Med ; 210(1): 157-72, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23296467

ABSTRACT

Colony-stimulating factor 1 (CSF1) and interleukin-34 (IL-34) are functional ligands of the CSF1 receptor (CSF1R) and thus are key regulators of the monocyte/macrophage lineage. We discovered that systemic administration of human recombinant CSF1 ameliorates memory deficits in a transgenic mouse model of Alzheimer's disease. CSF1 and IL-34 strongly reduced excitotoxin-induced neuronal cell loss and gliosis in wild-type mice when administered systemically before or up to 6 h after injury. These effects were accompanied by maintenance of cAMP responsive element-binding protein (CREB) signaling in neurons rather than in microglia. Using lineage-tracing experiments, we discovered that a small number of neurons in the hippocampus and cortex express CSF1R under physiological conditions and that kainic acid-induced excitotoxic injury results in a profound increase in neuronal receptor expression. Selective deletion of CSF1R in forebrain neurons in mice exacerbated excitotoxin-induced death and neurodegeneration. We conclude that CSF1 and IL-34 provide powerful neuroprotective and survival signals in brain injury and neurodegeneration involving CSF1R expression on neurons.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Neurons/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Base Sequence , Cell Survival , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Humans , Interleukins/genetics , Interleukins/pharmacology , Kainic Acid/toxicity , Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation , Prosencephalon/metabolism , Prosencephalon/pathology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction
11.
Nature ; 477(7362): 90-4, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21886162

ABSTRACT

In the central nervous system, ageing results in a precipitous decline in adult neural stem/progenitor cells and neurogenesis, with concomitant impairments in cognitive functions. Interestingly, such impairments can be ameliorated through systemic perturbations such as exercise. Here, using heterochronic parabiosis we show that blood-borne factors present in the systemic milieu can inhibit or promote adult neurogenesis in an age-dependent fashion in mice. Accordingly, exposing a young mouse to an old systemic environment or to plasma from old mice decreased synaptic plasticity, and impaired contextual fear conditioning and spatial learning and memory. We identify chemokines--including CCL11 (also known as eotaxin)--the plasma levels of which correlate with reduced neurogenesis in heterochronic parabionts and aged mice, and the levels of which are increased in the plasma and cerebrospinal fluid of healthy ageing humans. Lastly, increasing peripheral CCL11 chemokine levels in vivo in young mice decreased adult neurogenesis and impaired learning and memory. Together our data indicate that the decline in neurogenesis and cognitive impairments observed during ageing can be in part attributed to changes in blood-borne factors.


Subject(s)
Chemokines/blood , Chemokines/metabolism , Learning/physiology , Neurogenesis/physiology , Aging , Animals , Chemokine CCL11/blood , Chemokine CCL11/cerebrospinal fluid , Chemokine CCL11/metabolism , Chemokine CCL11/pharmacology , Chemokines/cerebrospinal fluid , Female , Learning/drug effects , Learning Disabilities/blood , Learning Disabilities/cerebrospinal fluid , Learning Disabilities/physiopathology , Male , Memory Disorders/blood , Memory Disorders/cerebrospinal fluid , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Parabiosis , Plasma/chemistry , Time Factors
12.
J Clin Invest ; 120(8): 2782-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628203

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) is a key hormonal system regulating blood pressure. However, expression of RAAS components has recently been detected in immune cells, and the RAAS has been implicated in several mouse models of autoimmune disease. Here, we have identified Ang II as a paracrine mediator, sustaining inflammation in the CNS in the EAE mouse model of MS via TGF-beta. Ang II type 1 receptors (AT1Rs) were found to be primarily expressed in CNS-resident cells during EAE. In vitro, astrocytes and microglia responded to Ang II treatment by inducing TGF-beta expression via a pathway involving the TGF-beta-activating protease thrombospondin-1 (TSP-1). TGF-beta upregulation in astrocytes and microglia during EAE was blocked with candesartan (CA), an inhibitor of AT1R. Treatment of EAE with CA ameliorated paralysis and blunted lymphocyte infiltration into the CNS, outcomes that were also seen with genetic ablation of AT1Ra and treatment with an inhibitor of TSP-1. These data suggest that AT1R antagonists, frequently prescribed as antihypertensives, may be useful to interrupt this proinflammatory, CNS-specific pathway in individuals with MS.


Subject(s)
Angiotensin II/physiology , Encephalomyelitis, Autoimmune, Experimental/etiology , Transforming Growth Factor beta/physiology , Animals , Benzimidazoles/therapeutic use , Biphenyl Compounds , Cells, Cultured , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Mice , Mice, Inbred C57BL , Receptor, Angiotensin, Type 1/analysis , Renin-Angiotensin System/physiology , Signal Transduction , Tetrazoles/therapeutic use , Thrombospondin 1/analysis , Transforming Growth Factor beta/analysis
13.
Nat Immunol ; 7(12): 1317-25, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086188

ABSTRACT

Transcriptional programs that initiate and sustain the proliferation, differentiation and survival of CD8(+) T cells during immune responses are not completely understood. Here we show that inhibitor of DNA binding 2 (Id2), an antagonist of E protein transcription factors, was upregulated in CD8(+) T cells during infection and that expression of Id2 was maintained in memory CD8(+) T cells. Although Id2-deficient naive CD8(+) T cells recognized antigen and proliferated normally early after infection, effector CD8(+) T cells did not accumulate because the cells were highly susceptible to apoptosis. Id2-deficient CD8(+) T cells responding to infection had changes in the expression of genes that influence survival and had altered memory formation. Our data emphasize the importance of Id2 in regulating gene expression by CD8(+) T cells and the magnitude of effector responses, suggesting a mechanism involving Id protein- and E protein-mediated survival and differentiation of mature T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Inhibitor of Differentiation Protein 2/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Gene Expression/immunology , Inhibitor of Differentiation Protein 2/metabolism , Listeriosis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...