Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Pathogens ; 12(8)2023 Jul 31.
Article En | MEDLINE | ID: mdl-37623960

Streptococcus pyogenes (Group A Streptococcus, GAS) bacteria cause a spectrum of human diseases ranging from self-limiting pharyngitis and mild, uncomplicated skin infections (impetigo, erysipelas, and cellulitis) to highly morbid and rapidly invasive, life-threatening infections such as streptococcal toxic shock syndrome and necrotizing fasciitis (NF). HLA class II allelic polymorphisms are linked with differential outcomes and severity of GAS infections. The dysregulated immune response and peripheral cytokine storm elicited due to invasive GAS infections increase the risk for toxic shock and multiple organ failure in genetically susceptible individuals. We hypothesized that, while the host immune mediators regulate the immune responses against peripheral GAS infections, these interactions may simultaneously trigger neuropathology and, in some cases, induce persistent alterations in the glial phenotypes. Here, we studied the consequences of peripheral GAS skin infection on the brain in an HLA-II transgenic mouse model of GAS NF with and without treatment with an antibiotic, clindamycin (CLN). Mice expressing the human HLA-II DR3 (DR3) or the HLA-II DR4 (DR4) allele were divided into three groups: (i) uninfected controls, (ii) subcutaneously infected with a clinical GAS strain isolated from a patient with GAS NF, and (iii) GAS-infected with CLN treatment (10 mg/kg/5 days, intraperitoneal). The groups were monitored for 15 days post-infection. Skin GAS burden and lesion area, splenic and hippocampal mRNA levels of inflammatory markers, and immunohistochemical changes in hippocampal GFAP and Iba-1 immunoreactivity were assessed. Skin GAS burden and hippocampal mRNA levels of the inflammatory markers S100A8/A9, IL-1ß, IL-33, inflammasome-related caspase-1 (Casp1), and NLRP6 were elevated in infected DR3 but not DR4 mice. The levels of these markers were significantly reduced following CLN treatment in DR3 mice. Although GAS was not detectable in the brain, astrocyte (GFAP) and microglia (Iba-1) activation were evident from increased GFAP and Iba-1 mRNA levels in DR3 and DR4 mice. However, CLN treatment significantly reduced GFAP mRNA levels in DR3 mice, not DR4 mice. Our data suggest a skin-brain axis during GAS NF, demonstrating that peripherally induced pathological conditions regulate neuroimmune changes and gliotic events in the brain.

2.
Int J Mol Sci ; 24(14)2023 Jul 17.
Article En | MEDLINE | ID: mdl-37511312

Alzheimer's disease (AD) is characterized by progressive cognitive decline and is a leading cause of death in the United States. Neuroinflammation has been implicated in the progression of AD, and several recent studies suggest that peripheral immune dysfunction may influence the disease. Continuing evidence indicates that intestinal dysbiosis is an attribute of AD, and inflammatory bowel disease (IBD) has been shown to aggravate cognitive impairment. Previously, we separately demonstrated that an IBD-like condition exacerbates AD-related changes in the brains of the AppNL-G-F mouse model of AD, while probiotic intervention has an attenuating effect. In this study, we investigated the combination of a dietary probiotic and an IBD-like condition for effects on the brains of mice. Male C57BL/6 wild type (WT) and AppNL-G-F mice were randomly divided into four groups: vehicle control, oral probiotic, dextran sulfate sodium (DSS), and DSS given with probiotics. As anticipated, probiotic treatment attenuated the DSS-induced colitis disease activity index in WT and AppNL-G-F mice. Although probiotic feeding significantly attenuated the DSS-mediated increase in WT colonic lipocalin levels, it was less protective in the AppNL-G-F DSS-treated group. In parallel with the intestinal changes, combined probiotic and DSS treatment increased microglial, neutrophil elastase, and 5hmC immunoreactivity while decreasing c-Fos staining compared to DSS treatment alone in the brains of WT mice. Although less abundant, probiotic combined with DSS treatment demonstrated a few similar changes in AppNL-G-F brains with increased microglial and decreased c-Fos immunoreactivity in addition to a slight increase in Aß plaque staining. Both probiotic and DSS treatment also altered the levels of several cytokines in WT and AppNL-G-F brains, with a unique increase in the levels of TNFα and IL-2 being observed in only AppNL-G-F mice following combined DSS and probiotic treatment. Our data indicate that, while dietary probiotic intervention provides protection against the colitis-like condition, it also influences numerous glial, cytokine, and neuronal changes in the brain that may regulate brain function and the progression of AD.


Alzheimer Disease , Colitis , Inflammatory Bowel Diseases , Mobile Applications , Probiotics , Mice , Male , Animals , Alzheimer Disease/therapy , Alzheimer Disease/etiology , Amyloid beta-Peptides , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/therapy , Colitis/complications , Inflammatory Bowel Diseases/complications , Cytokines , Probiotics/pharmacology , Probiotics/therapeutic use , Disease Models, Animal , Mice, Transgenic
3.
Curr Alzheimer Res ; 19(5): 335-350, 2022.
Article En | MEDLINE | ID: mdl-35718965

BACKGROUND: There is a well-described mechanism of communication between the brain and gastrointestinal system in which both organs influence the function of the other. This bi-directional communication suggests that disease in either organ may affect function in the other. OBJECTIVE: To assess whether the evidence supports gastrointestinal system inflammatory or degenerative pathophysiology as a characteristic of Alzheimer's disease (AD). METHODS: A review of both rodent and human studies implicating gastrointestinal changes in AD was performed. RESULTS: Numerous studies indicate that AD changes are not unique to the brain but also occur at various levels of the gastrointestinal tract involving both immune and neuronal changes. In addition, it appears that numerous conditions and diseases affecting regions of the tract may communicate to the brain to influence disease. CONCLUSION: Gastrointestinal changes represent an overlooked aspect of AD, representing a more system influence of this disease.


Alzheimer Disease , Brain , Gastrointestinal Tract , Humans , Neurons
4.
J Alzheimers Dis ; 80(2): 761-774, 2021.
Article En | MEDLINE | ID: mdl-33554902

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-ß (Aß) plaques, neuroinflammation, and neuronal death. There are several well-established genetic and environmental factors hypothesized to contribute to AD progression including air pollution. However, the molecular mechanisms by which air pollution exacerbates AD are unclear. OBJECTIVE: This study explored the effects of particulate matter exposure on AD-related brain changes using the APP/PS1 transgenic model of disease. METHODS: Male C57BL/6;C3H wild type and APP/PS1 mice were exposed to either filtered air (FA) or particulate matter sized under 2.5µm (PM2.5) for 6 h/day, 5 days/week for 3 months and brains were collected. Immunohistochemistry for Aß, GFAP, Iba1, and CD68 and western blot analysis for PS1, BACE, APP, GFAP, and Iba1 were performed. Aß ELISAs and cytokine arrays were performed on frozen hippocampal and cortical lysates, respectively. RESULTS: The Aß plaque load was significantly increased in the hippocampus of PM2.5-exposed APP/PS1 mice compared to their respective FA controls. Additionally, in the PM2.5-exposed APP/PS1 group, increased astrocytosis and microgliosis were observed as indicated by elevated GFAP, Iba1, and CD68 immunoreactivities. PM2.5 exposure also led to an elevation in the levels of PS1 and BACE in APP/PS1 mice. The cytokines TNF-α, IL-6, IL-1ß, IFN-γ, and MIP-3α were also elevated in the cortices of PM2.5-exposed APP/PS1 mice compared to FA controls. CONCLUSION: Our data suggest that chronic particulate matter exposure exacerbates AD by increasing Aß plaque load, gliosis, and the brain inflammatory status.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cytokines/metabolism , Gliosis , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Gliosis/metabolism , Gliosis/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/pathology
5.
J Alzheimers Dis ; 79(3): 1235-1255, 2021.
Article En | MEDLINE | ID: mdl-33427741

BACKGROUND: Although it is known that the brain communicates with the gastrointestinal (GI) tract via the well-established gut-brain axis, the influence exerted by chronic intestinal inflammation on brain changes in Alzheimer's disease (AD) is not fully understood. We hypothesized that increased gut inflammation would alter brain pathology of a mouse model of AD. OBJECTIVE: Determine whether colitis exacerbates AD-related brain changes. METHODS: To test this idea, 2% dextran sulfate sodium (DSS) was dissolved in the drinking water and fed ad libitum to male C57BL/6 wild type and AppNL-G-F mice at 6-10 months of age for two cycles of three days each. DSS is a negatively charged sulfated polysaccharide which results in bloody diarrhea and weight loss, changes similar to human inflammatory bowel disease (IBD). RESULTS: Both wild type and AppNL-G-F mice developed an IBD-like condition. Brain histologic and biochemical assessments demonstrated increased insoluble Aß1-40/42 levels along with the decreased microglial CD68 immunoreactivity in DSS treated AppNL-G-F mice compared to vehicle treated AppNL-G-F mice. CONCLUSION: These data demonstrate that intestinal dysfunction is capable of altering plaque deposition and glial immunoreactivity in the brain. This study increases our knowledge of the impact of peripheral inflammation on Aß deposition via an IBD-like model system.


Colitis/complications , Dextran Sulfate/pharmacology , Inflammation/complications , Plaque, Amyloid/etiology , Animals , Blotting, Western , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/pathology
6.
Front Cell Neurosci ; 14: 200, 2020.
Article En | MEDLINE | ID: mdl-32719587

Aging is a major risk factor for Alzheimer's disease (AD). Insulin-like growth factor-1 receptor (IGF-1R) regulates general aging and lifespan. However, the contribution of IGF-1 to age-related AD pathology and progression is highly controversial. Based on our previous work, AßPP/PS1 double transgenic mice, which express human mutant amyloid precursor protein (APP) and presenilin-1 (PS-1), demonstrated a decrease in brain IGF-1 levels when they were crossed with IGF-1 deficient Ames dwarf mice (df/df). Subsequently, a reduction in gliosis, amyloid-ß (Aß) plaque deposition, and Aß1-40/42 concentrations were observed in this mouse model. This supported the hypothesis that IGF-1 may contribute to the progression of the disease. To assess the role of IGF-1 in AD, 9-10-month-old male littermate control wild type and AßPP/PS1 mice were randomly divided into two treatment groups including control vehicle (DMSO) and picropodophyllin (PPP), a selective, competitive, and reversible IGF-1R inhibitor. The brain penetrant inhibitor was given ip. at 1 mg/kg/day. Mice were sacrificed after 7 days of daily injection and the brains, spleens, and livers were collected to quantify histologic and biochemical changes. The PPP-treated AßPP/PS1 mice demonstrated attenuated insoluble Aß1-40/42. Additionally, an attenuation in microgliosis and protein p-tyrosine levels was observed due to drug treatment in the hippocampus. Our data suggest IGF-1R signaling is associated with disease progression in this mouse model. More importantly, modulation of the brain IGF-1R signaling pathway, even at mid-life, was enough to attenuate aspects of the disease phenotype. This suggests that small molecule therapy targeting the IGF-1R pathway may be viable for late-stage disease treatment.

7.
J Alzheimers Dis ; 76(3): 1083-1102, 2020.
Article En | MEDLINE | ID: mdl-32623399

BACKGROUND: The intestinal microbiota and its metabolites, particularly short-chain fatty acids (SCFAs), have been implicated in immune function, host metabolism, and even behavior. OBJECTIVE: This study was performed to investigate whether probiotic administration influences levels of intestinal microbiota and their metabolites in a fashion that may attenuate brain changes in a mouse model of Alzheimer's disease (AD). METHODS: C57BL/6 wild-type (WT) mice were compared to AppNL-G-Fmice. The animals were treated with either vehicle or probiotic (VSL#3) for 8 weeks. Fecal microbiome analysis along with Aß, GFAP, Iba-1, c-Fos, and Ki-67 immunohistochemistry was done. SCFAs were analyzed in serum and brains using UPLC-MS/MS. RESULTS: Probiotic (VSL#3) supplementation for 2 months resulted in altered microbiota in both WT and AppNL-G-Fmice. An increase in serum SCFAs acetate, butyrate, and lactate were found in both genotypes following VSL#3 treatment. Propionate and isobutyrate were only increased in AppNL-G-Fmice. Surprisingly, VSL#3 only increased lactate and acetate in brains of AppNL-G-Fmice. No significant differences were observed between vehicle and VSL#3 fed AppNL-G-Fhippocampal immunoreactivities of Aß, GFAP, Iba-1, and Ki-67. However, hippocampal c-Fos staining increased in VSL#3 fed AppNL-G-Fmice. CONCLUSION: These data demonstrate intestinal dysbiosis in the AppNL-G-Fmouse model of AD. Probiotic VSL#3 feeding altered both serum and brain levels of lactate and acetate in AppNL-G-Fmice correlating with increased expression of the neuronal activity marker, c-Fos.


Alzheimer Disease/drug therapy , Butyrates/pharmacology , Fatty Acids, Volatile/metabolism , Probiotics/pharmacology , Alzheimer Disease/chemically induced , Animals , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Mice, Transgenic , Microbiota/drug effects
8.
Curr Alzheimer Res ; 17(12): 1133-1144, 2020.
Article En | MEDLINE | ID: mdl-33463464

BACKGROUND: Beta amyloid (Aß) peptide containing plaque aggregations in the brain are a hallmark of Alzheimer's Disease (AD). However, Aß is produced by cell types outside of the brain suggesting that the peptide may serve a broad physiologic purpose. OBJECTIVE: Based upon our prior work documenting expression of amyloid ß precursor protein (APP) in intestinal epithelium we hypothesized that salivary epithelium might also express APP and be a source of Aß. METHODS: To begin testing this idea, we compared human age-matched control and AD salivary glands to C57BL/6 wild type, AppNL-G-F , and APP/PS1 mice. RESULTS: Both male and female AD, AppNL-G-F , and APP/PS1 glands demonstrated robust APP and Aß immunoreactivity. Female AppNL-G-F mice had significantly higher levels of pilocarpine stimulated Aß 1-42 compared to both wild type and APP/PS1 mice. No differences in male salivary Aß levels were detected. No significant differences in total pilocarpine stimulated saliva volumes were observed in any group. Both male and female AppNL-G-F but not APP/PS1 mice demonstrated significant differences in oral microbiome phylum and genus abundance compared to wild type mice. Male, but not female, APP/PS1 and AppNL-G-F mice had significantly thinner molar enamel compared to their wild type counterparts. CONCLUSION: These data support the idea that oral microbiome changes exist during AD in addition to changes in salivary Aß and oral health.


Amyloid beta-Peptides/metabolism , Brain/metabolism , Disease Models, Animal , Microbiota , Plaque, Amyloid/metabolism , Saliva/metabolism , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL
9.
MethodsX ; 6: 2028-2036, 2019.
Article En | MEDLINE | ID: mdl-31667100

The appropriate methodological approach for intestinal preparation enables researchers to create representative histological and immunostaining images which validate their biochemical data. The Swiss-roll technique was first introduced by Reilly and Kirsner in 1965. Later, Moolenbeek and Ruitenberg described a detailed procedure to longitudinally study the rodent intestine in 1981 [1]. In this publication, our slightly different approach for co-embedding four different Swiss-rolls in a gelatin block provides a full-length overview of cross-sectional colons on a single slide. This protocol allows for longitudinal histologic examination of multiple tissue samples on a single slide simultaneously. In this method, antigenicity is retained for immunohistology. In addition, the accessibility of samples during the prolonged hardening time required of the gelatin matrix allows the tissue samples to be adjusted/re-adjusted to provide the desired orientation and spatial arrangement for ideal cross sections with similar planes of section and optimum space utilization for slide mounting. Although not the focus of this protocol, the room temperature stability of the gelatin matrix and the ability to contain numerous tissue samples in a block allows the flexibility of performing thicker sections for free-floating tissue staining and ease of mounting a single gelatin sheet rather than individual tissue sections. This is a convenient approach for allowing precise preparation of multi-tissue blocks and simultaneous sectioning, staining, and slide mounting of tissue for subsequent comparisons. •A single gelatin block is prepared by simultaneously embedding at least four different intestinal Swiss-rolls.•The tissue orientation can be adjusted for each sample as desired which facilitates the comparison of different colon samples on a single gelatin section.•The gelatin sections containing tissue samples are stable at least overnight at room temperature for staining.

10.
Front Cell Neurosci ; 13: 320, 2019.
Article En | MEDLINE | ID: mdl-31379506

Etiology of neuropsychiatric disorders is complex, involving multiple factors that can affect the type and severity of symptoms. Although precise causes are far from being identified, allergy or other forms of hypersensitivity to dietary ingredients have been implicated in triggering or worsening of behavioral and emotional symptoms, especially in patients suffering from depression, anxiety, attention-deficit hyperactivity, and/or autism. Among such ingredients, cow's milk, along with wheat gluten, is commonly suspected. However, the contributory role of cow's milk in these disorders has not been elucidated due to insufficient pathophysiological evidence. In the present study, we therefore investigated neuroinflammatory changes that are associated with behavioral abnormality using a non-anaphylactic mouse model of cow's milk allergy (CMA). Male and female C57BL/6J mice were subjected to a 5-week oral sensitization procedure without or with a major milk allergen, beta-lactoglobulin (BLG). All mice were then later challenged with BLG, and their anxiety- and depression-associated behaviors were subsequently assessed during the 6th and 7th weeks. We found that BLG-sensitized male mice exhibited significantly increased anxiety- and depression-like behavior, although they did not display anaphylactic reactions when challenged with BLG. Female behavior was not noticeably affected by BLG sensitization. Upon examination of the small intestines, reduced immunoreactivity to occludin was detected in the ileal mucosa of BLG-sensitized mice although the transcriptional expression of this tight-junction protein was not significantly altered when measured by quantitative RT-PCR. On the other hand, the expression of tumor necrosis factor alpha (TNFα) in the ileal mucosa was significantly elevated in BLG-sensitized mice, suggesting the sensitization had resulted in intestinal inflammation. Inflammatory responses were also detected in the brain of BLG-sensitized mice, determined by the hypertrophic morphology of GFAP-immunoreactive astrocytes. These reactive astrocytes were particularly evident near the blood vessels in the midbrain region, resembling the perivascular barrier previously reported by others in experimental autoimmune encephalitis (EAE) mouse models. Interestingly, increased levels of COX-2 and TNFα were also found in this region. Taken together, our results demonstrated that BLG sensitization elicits inflammatory responses in the intestine and brain without overt anaphylactic signs of milk allergy, signifying food allergy as a potential pathogenic factor of neuropsychiatric disorders.

11.
Neurobiol Aging ; 81: 166-176, 2019 09.
Article En | MEDLINE | ID: mdl-31284126

The amyloid beta (Aß) peptide is associated with the neurodegenerative and inflammatory changes in brains affected by Alzheimer's disease (AD). We hypothesized that the enteric nervous system also produces Aß in an intestinal component of disease. To test this idea, we compared C57BL/6 wild-type (WT) male and female mice to two models of Alzheimer's disease, amyloid precursor protein (APP)/presenilin 1 (PS1) mice and amyloid precursor protein NL-G-F (AppNL-G-F) mice, at 3, 6, and 12 months of age. Brain Aß plaque deposition in AppNL-G-F mice preceded that in the APP/PS1 mice, observable by 3 months. Three-month-old female AppNL-G-F mice had decreased intestinal motility compared with WT and APP/PS1 mice. However, 3-month-old female APP/PS1 mice demonstrated increased intestinal permeability compared with WT and AppNL-G-F mice. Both sexes of APP/PS1 and AppNL-G-F mice demonstrated increased colon lipocalin 2 mRNA and insoluble Aß 1-42 levels at 3 months. These data demonstrate an unrecognized enteric aspect of disease in 2 different mouse models correlating with the earliest brain changes.


Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Intestinal Mucosa/metabolism , Temporal Lobe/metabolism , Amyloid beta-Protein Precursor , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Gastrointestinal Motility , Intestines/innervation , Lipocalin-2/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1
12.
J Neurochem ; 149(5): 562-581, 2019 06.
Article En | MEDLINE | ID: mdl-30702751

This review discusses the profound connection between microglia, neuroinflammation, and Alzheimer's disease (AD). Theories have been postulated, tested, and modified over several decades. The findings have further bolstered the belief that microglia-mediated inflammation is both a product and contributor to AD pathology and progression. Distinct microglia phenotypes and their function, microglial recognition and response to protein aggregates in AD, and the overall role of microglia in AD are areas that have received considerable research attention and yielded significant results. The following article provides a historical perspective of microglia, a detailed discussion of multiple microglia phenotypes including dark microglia, and a review of a number of areas where microglia intersect with AD and other pathological neurological processes. The overall breadth of important discoveries achieved in these areas significantly strengthens the hypothesis that neuroinflammation plays a key role in AD. Future determination of the exact mechanisms by which microglia respond to, and attempt to mitigate, protein aggregation in AD may lead to new therapeutic strategies.


Alzheimer Disease/immunology , Inflammation/immunology , Microglia/immunology , Nerve Degeneration/immunology , Alzheimer Disease/pathology , Animals , Humans , Microglia/metabolism , Nerve Degeneration/pathology
13.
Circ Res ; 124(1): 142-149, 2019 01 04.
Article En | MEDLINE | ID: mdl-30605407

In a somewhat narrow diagnostic lens, Alzheimer disease (AD) has been considered a brain-specific disease characterized by the presence of Aß (ß-amyloid) plaques and tau neural fibrillary tangles and neural inflammation; these pathologies lead to neuronal death and consequently clinical symptoms, such as memory loss, confusion, and impaired cognitive function. However, for decades, researchers have noticed a link between various cardiovascular abnormalities and AD-such as heart failure, coronary artery disease, atrial fibrillation, and vasculopathy. A considerable volume of work has pointed at this head to heart connection, focusing mainly on associations between cerebral hypoperfusion and neuronal degradation. However, new evidence of a possible systemic or metastatic profile to AD calls for further analysis of this connection. Aß aggregations-biochemically and structurally akin to those found in the typical AD pathology-are now known to be present in the hearts of individuals with idiopathic dilated cardiomyopathy, as well as the hearts of patients with AD. These findings suggest a potential systemic profile of proteinopathies and a new hypothesis for the link between peripheral and central symptoms of heart failure and AD. Herein, we provide an overview of the cardiovascular links to Alzheimer disease.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Plaque, Amyloid , Alzheimer Disease/epidemiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain/pathology , Brain/physiopathology , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Cerebrovascular Circulation , Humans , Inflammation Mediators/metabolism , Oxidative Stress , Prognosis , Reactive Oxygen Species/metabolism , Risk Factors , Signal Transduction
14.
Am J Physiol Endocrinol Metab ; 316(1): E106-E120, 2019 01 01.
Article En | MEDLINE | ID: mdl-30422705

The amyloid precursor protein (APP) is a type I transmembrane glycoprotein widely studied for its role as the source of ß-amyloid peptide, accumulation of which is causal in at least some cases of Alzheimer's disease (AD). APP is expressed ubiquitously and is involved in diverse biological processes. Growing bodies of evidence indicate connections between AD and somatic metabolic disorders related to type 2 diabetes, and App-/- mice show alterations in glycemic regulation. We find that App-/- mice have higher levels of insulin-degrading enzyme (IDE) mRNA, protein, and activity compared with wild-type controls. This regulation of IDE by APP was widespread across numerous tissues, including liver, skeletal muscle, and brain as well as cell types within neural tissue, including neurons, astrocytes, and microglia. RNA interference-mediated knockdown of APP in the SIM-A9 microglia cell line elevated IDE levels. Fasting levels of blood insulin were lower in App-/- than App+/+ mice, but the former showed a larger increase in response to glucose. These low basal levels may enhance peripheral insulin sensitivity, as App-/- mice failed to develop impairment of glucose tolerance on a high-fat, high-sucrose ("Western") diet. Insulin levels and insulin signaling were also lower in the App-/- brain; synaptosomes prepared from App-/- hippocampus showed diminished insulin receptor phosphorylation compared with App+/+ mice when stimulated ex vivo. These findings represent a new molecular link connecting APP to metabolic homeostasis and demonstrate a novel role for APP as an upstream regulator of IDE in vivo.


Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Insulin Resistance/genetics , Insulin/metabolism , Insulysin/genetics , Liver/metabolism , Muscle, Skeletal/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Cell Line , Diet, High-Fat , Diet, Western , Glucose Intolerance/genetics , Hippocampus/metabolism , Insulysin/metabolism , Mice , Mice, Knockout , Microglia/metabolism , Neurons/metabolism , Phosphorylation , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Synaptosomes/metabolism
15.
Environ Pollut ; 241: 279-288, 2018 Oct.
Article En | MEDLINE | ID: mdl-29843010

Environmental exposure to air pollution has been linked to a number of health problems including organ rejection, lung damage and inflammation. While the deleterious effects of air pollution in adult animals are well documented, the long-term consequences of particulate matter (PM) exposure during animal development are uncertain. In this study we tested the hypothesis that environmental exposure to PM 2.5 µm in diameter in utero promotes long term inflammation and neurodegeneration. We evaluated the behavior of PM exposed animals using several tests and observed deficits in spatial memory without robust changes in anxiety-like behavior. We then examined how this affects the brains of adult animals by examining proteins implicated in neurodegeneration, synapse formation and inflammation by western blot, ELISA and immunohistochemistry. These tests revealed significantly increased levels of COX2 protein in PM2.5 exposed animal brains in addition to changes in synaptophysin and Arg1 proteins. Exposure to PM2.5 also increased the immunoreactivity for GFAP, a marker of activated astrocytes. Cytokine concentrations in the brain and spleen were also altered by PM2.5 exposure. These findings indicate that in utero exposure to particulate matter has long term consequences which may affect the development of both the brain and the immune system in addition to promoting inflammatory change in adult animals.


Air Pollutants/toxicity , Nervous System/immunology , Particulate Matter/toxicity , Toxicity Tests , Adult , Air Pollutants/analysis , Air Pollution/analysis , Animals , Anxiety/chemically induced , Behavior, Animal/drug effects , Biomarkers/analysis , Brain/drug effects , Environmental Exposure/analysis , Humans , Male , Mice , Particulate Matter/analysis , Phenotype
16.
J Neuroinflammation ; 15(1): 120, 2018 Apr 23.
Article En | MEDLINE | ID: mdl-29685134

BACKGROUND: Growing evidence has strengthened the association of food allergy with neuropsychiatric symptoms such as depression, anxiety, and autism. However, underlying mechanisms by which peripheral allergic responses lead to behavioral dysfunction are yet to be determined. Allergen-activated mast cells may serve as mediators by releasing histamine and other inflammatory factors that could adversely affect brain function. We hypothesized that eliciting food allergy in experimental animals would result in behavioral changes accompanied by mast cell accumulation in the brain. Our hypothesis was tested in a mouse model of milk allergy using bovine milk whey proteins (WP) as the allergen. METHODS: Male and female C57BL/6 mice at 4 weeks (young) and 10 months (old) of age underwent 5-week WP sensitization with weekly intragastric administration of 20 mg WP and 10 µg cholera toxin as an adjuvant. Age-matched sham animals were given the vehicle containing only the adjuvant. All animals were orally challenged with 50 mg WP in week 6 and their intrinsic digging behavior was assessed the next day. Animals were sacrificed 3 days after the challenge, and WP-specific serum IgE, intestinal and brain mast cells, glial activation, and epigenetic DNA modification in the brain were examined. RESULTS: WP-sensitized males showed significantly less digging activity than the sham males in both age groups while no apparent difference was observed in females. Mast cells and their activities were evident in the intestines in an age- and sex-dependent manner. Brain mast cells were predominantly located in the region between the lateral midbrain and medial hippocampus, and their number increased in the WP-sensitized young, but not old, male brains. Noticeable differences in for 5-hydroxymethylcytosine immunoreactivity were observed in WP mice of both age groups in the amygdala, suggesting epigenetic regulation. Increased microglial Iba1 immunoreactivity and perivascular astrocytes hypertrophy were also observed in the WP-sensitized old male mice. CONCLUSIONS: Our results demonstrated that food allergy induced behavioral abnormality, increases in the number of mast cells, epigenetic DNA modification in the brain, microgliosis, and astrocyte hypertrophy in a sex- and age-dependent manner, providing a potential mechanism by which peripheral allergic responses evoke behavioral dysfunction.


Aging , Encephalitis/etiology , Food Hypersensitivity/complications , Food Hypersensitivity/etiology , Mast Cells/pathology , Mental Disorders/etiology , Whey Proteins/toxicity , Animals , Disease Models, Animal , Female , Immunoglobulin E/metabolism , Male , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Occludin/metabolism , RNA, Messenger/metabolism , Sex Factors , Tryptases/genetics , Tryptases/metabolism , Whey Proteins/immunology
17.
J Neurochem ; 143(6): 736-749, 2017 12.
Article En | MEDLINE | ID: mdl-28881033

Aggregation and accumulation of amyloid-ß peptide (Aß) is a key component of Alzheimer's disease (AD). While monomeric Aß appears to be benign, oligomers adopt a biologically detrimental structure. These soluble structures can be detected in AD brain tissue by antibodies that demonstrate selectivity for aggregated Aß. Protofibrils are a subset of soluble oligomeric Aß species and are described as small (< 100 nm) curvilinear assemblies enriched in ß-sheet structure. Our own in vitro studies demonstrate that microglial cells are much more sensitive to soluble Aß42 protofibrils compared to Aß42 monomer or insoluble Aß42 fibrils. Protofibrils interact with microglia, trigger Toll-like receptor signaling, elicit cytokine transcription and expression, and are rapidly taken up by the cells. Because of the importance of this Aß species, we sought to develop an antibody that selectively recognizes protofibrils over other Aß species. Immunization of rabbits with isolated Aß42 protofibrils generated a high-titer anti serum with a strong affinity for Aß42 protofibrils. The antiserum, termed AbSL, was selective for Aß42 protofibrils over Aß42 monomers and Aß42 fibrils. AbSL did not react with amyloid precursor protein and recognized distinct pathological features in AD transgenic mouse brain slices. Competition studies with an Aß antibody that targets residues 1-16 indicated that the conformational epitope for AbSL involved the N-terminal region of protofibrils in some manner. The newly developed antibody may have potential diagnostic and therapeutic uses in AD tissue and patients, and targeting of protofibrils in AD may have beneficial effects. Read the Editorial Highlight for this article on page 621. Cover Image for this issue: doi. 10.1111/jnc.13827.


Alzheimer Disease , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Animals , Antibody Specificity , Epitopes , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Conformation, beta-Strand
18.
J Endocrinol ; 235(1): 49-67, 2017 Oct.
Article En | MEDLINE | ID: mdl-28710249

The amyloid precursor protein (APP) has been extensively investigated for its role in the production of amyloid beta (Aß), a plaque-forming peptide in Alzheimer's disease (AD). Epidemiological evidence suggests type 2 diabetes is a risk factor for AD. The pancreas is an essential regulator of blood glucose levels through the secretion of the hormones insulin and glucagon. Pancreatic dysfunction is a well-characterized consequence of type 1 and type 2 diabetes. In this study, we have examined the expression and processing of pancreatic APP to test the hypothesis that APP may play a role in pancreatic function and the pathophysiology of diabetes. Our data demonstrate the presence of APP within the pancreas, including pancreatic islets in both mouse and human samples. Additionally, we report that the APP/PS1 mouse model of AD overexpresses APP within pancreatic islets, although this did not result in detectable levels of Aß. We compared whole pancreas and islet culture lysates by Western blot from C57BL/6 (WT), APP-/- and APP/PS1 mice and observed APP-dependent differences in the total protein levels of GLUT4, IDE and BACE2. Immunohistochemistry for BACE2 detected high levels in pancreatic α cells. Additionally, both mouse and human islets processed APP to release sAPP into cell culture media. Moreover, sAPP stimulated insulin but not glucagon secretion from islet cultures. We conclude that APP and its metabolites are capable of influencing the basic physiology of the pancreas, possibly through the release of sAPP acting in an autocrine or paracrine manner.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Female , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Precursors/genetics , Protein Precursors/metabolism
19.
J Alzheimers Dis ; 58(3): 775-787, 2017.
Article En | MEDLINE | ID: mdl-28505967

Alzheimer's disease (AD) brains are characterized by fibrillar amyloid-ß (Aß) peptide containing plaques and associated reactive microglia. The proinflammatory phenotype of the microglia suggests that they may negatively affect disease course and contribute to behavioral decline. This hypothesis predicts that attenuating microglial activation may provide benefit against disease. Prior work from our laboratory and others has characterized a role for the transcription factor, nuclear factor of activated T cells (NFAT), in regulating microglial phenotype in response to different stimuli, including Aß peptide. We observed that the NFATc2 isoform was the most highly expressed in murine microglia cultures, and inhibition or deletion of NFATc2 was sufficient to attenuate the ability of the microglia to secrete cytokines. In order to determine whether the NFATc2 isoform, in particular, was a valid immunomodulatory target in vivo, we crossed an NFATc2-/- line to a well-known AD mouse model, an AßPP/PS1 mouse line. As expected, the AßPP/PS1 x NFATc2-/- mice had attenuated cytokine levels compared to AßPP/PS1 mice as well as reduced microgliosis and astrogliosis with no effect on plaque load. Although some species differences in relative isoform expression may exist between murine and human microglia, it appears that microglial NFAT activity is a viable target for modulating the proinflammatory changes that occur during AD.


Alzheimer Disease/metabolism , Microglia/metabolism , NFATC Transcription Factors/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Line , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Gliosis/metabolism , Gliosis/pathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , RNA, Messenger/metabolism
20.
Sci Rep ; 7: 43725, 2017 03 06.
Article En | MEDLINE | ID: mdl-28262782

It is well known that mutations in the gene coding for amyloid precursor protein are responsible for autosomal dominant forms of Alzheimer's disease. Proteolytic processing of the protein leads to a number of metabolites including the amyloid beta peptide. Although brain amyloid precursor protein expression and amyloid beta production are associated with the pathophysiology of Alzheimer's disease, it is clear that amyloid precursor protein is expressed in numerous cell types and tissues. Here we demonstrate that amyloid precursor protein is involved in regulating the phenotype of both adipocytes and peripheral macrophages and is required for high fat diet-dependent weight gain in mice. These data suggest that functions of this protein include modulation of the peripheral immune system and lipid metabolism. This biology may have relevance not only to the pathophysiology of Alzheimer's disease but also diet-associated obesity.


Amyloid beta-Protein Precursor/metabolism , Macrophages/metabolism , Phenotype , Weight Gain , Adipose Tissue/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/immunology , Animals , Biomarkers , Diet , Diet, High-Fat , Disease Models, Animal , Inflammation Mediators/metabolism , Macrophages/immunology , Male , Mice , Mice, Knockout , Weight Gain/genetics
...