Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Biochem Biophys Res Commun ; 638: 28-35, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36436339

ABSTRACT

Over the last decade, 64Cu-labeling of monoclonal antibody (mAb) via inverse electron demand Diels-Alder click chemistry (IEDDA) have received much attention. Despite the tetrazine-transcyclooctene (Tz-TCO) click chemistry's convenience and efficiency in mAb labeling, there is limited information about the ideal parameters in the development of click chemistry mediated (radio)immunoconjugates. This encourages us to conduct a systematic optimization while concurrently determining the physiochemical characteristics of the model mAb, trastuzumab, and TCO conjugates. To accomplish this, we investigated a few critical parameters, first, we determined the degree of conjugations with varying molar equivalents (eq.) of TCO (3, 5, 10, and 15 eq.). Through analytical techniques like size exclusion chromatography, dynamic light scattering, and zeta potential, qualitative analysis were performed to determine the purity, degree of aggregation and net charge of the conjugates. We found that as the degree of conjugation increased the purity of intact mAb fraction is compromised and net charge of conjugates became less positive. Next, all trastuzumab-PEG4-TCO conjugates with varying molar ratio and quantity (30, 50, 100, 200, 250 µg) were radiolabeled with 64Cu-NOTA-PEG4-Tz via IEDDA click chemistry and radiochemical yields were determined by radio-thin layer chromatography. The radiochemical yields of trastuzumab conjugates improved with increased amount and molar ratio. Next, we investigated the effect of the radioprotectant ascorbic acid (AA) of varied concentrations (0.25, 0.5, 0.75, 1 mM) on radiochemical yields and subsequent pharmacokinetics. A concentration of 0.25 mM of AA was found to be optimal for click reaction and in vivo biodistribution. Finally, we investigated the indirect influence of bioconjugation buffers on radiochemical yields and biodistribution in NIH3T6.7 tumor models that resulted approximately ∼11 %ID/g tumor uptake.


Subject(s)
Copper Radioisotopes , Neoplasms , Humans , Trastuzumab , Click Chemistry/methods , Tissue Distribution , Antibodies, Monoclonal , Radiopharmaceuticals/pharmacokinetics , Cell Line, Tumor
2.
Eur J Nucl Med Mol Imaging ; 49(12): 4073-4087, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35680737

ABSTRACT

PURPOSE: Hydrogen sulfide (H2S) plays important roles in brain pathophysiology. However, nuclear imaging probes for the in vivo detection of brain H2S in living animals have not been developed. Here, we report the first nuclear imaging probe that enables in vivo imaging of endogenous H2S in the brain of live mice. METHODS: Utilizing a bis(thiosemicarbazone) backbone, a fluorescent ATSM-FITC conjugate was synthesized. Its copper complex, Cu(ATSM-FITC) was thoroughly tested as a biosensor for H2S. The same ATSM-FITC ligand was quantitatively labeled with [64Cu]CuCl2 to obtain a radioactive [64Cu][Cu(ATSM-FITC)] imaging probe. Biodistribution and positron emission tomography (PET) imaging studies were performed in healthy mice and neuroinflammation models. RESULTS: The Cu(ATSM-FITC) complex reacts instantly with H2S to release CuS and becomes fluorescent. It showed excellent reactivity, sensitivity, and selectivity to H2S. Endogenous H2S levels in living cells were successfully detected by fluorescence microscopy. Exceptionally high brain uptake of [64Cu][Cu(ATSM-FITC)] (> 9% ID/g) was observed in biodistribution and PET imaging studies. Subtle changes in brain H2S concentrations in live mice were accurately detected by quantitative PET imaging. Due to its dual modality feature, increased H2S levels in neuroinflammation models were characterized at the subcellular level by fluorescence imaging and at the whole-body scale by PET imaging. CONCLUSION: Our biosensor can be readily utilized to study brain H2S function in live animal models and shows great potential as a novel imaging agent for diagnosing brain diseases.


Subject(s)
Coordination Complexes , Hydrogen Sulfide , Organometallic Compounds , Thiosemicarbazones , Animals , Brain/diagnostic imaging , Copper , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Ligands , Mice , Neuroinflammatory Diseases , Tissue Distribution
3.
Biochem Biophys Res Commun ; 479(2): 331-337, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27641666

ABSTRACT

Microglia-mediated clearance of amyloid beta-protein (Aß) via Toll-like receptor 4 (TLR4) signaling may play an important role in the pathogenesis of Alzheimer's disease (AD). However, as the disease progresses, activated microglia appear to become incapable of clearing Aß deposits. Because repeated exposure to a TLR4 ligand leads to a diminished response of monocytes/macrophages to lipopolysaccharide (LPS) and because aggregated Aß is a TLR4 ligand, we hypothesize that chronic exposure of microglia to Aß deposits may induce a state of Toll-like receptor (TLR) signaling dysfunction, leading to decreased Aß clearance and accelerated disease progression. LPS or phosphate-buffered saline (PBS) was injected into the hippocampus of AD-model (TgAPP/PS1) and wild-type (non-Tg) mice before and after the onset of Aß deposition, at age 2 and 12 months, respectively. Brain specimens were collected 7 days post-injection and analyzed for microglial activation and Aß load. While LPS-injected 2-month-old non-Tg mice showed 48-fold and 11-fold greater Iba1 immunoreactivity in the neocortex and hippocampus, respectively, compared with PBS-injected mice, LPS-injected 2-month-old TgAPP/PS1 mice had 61-fold and 13-fold increases in the neocortex and hippocampus, respectively. LPS injection activated microglia more strongly in TgAPP/PS1 mice than in non-Tg mice at 2 months of age. In contrast, at 12 months of age, Iba1 immunoreactivity of microglia was increased 541-fold and 38-fold in the neocortex and hippocampus, respectively, in LPS-injected non-Tg mice and 2.7-fold and 3.3-fold in the neocortex and hippocampus, respectively, in LPS-injected TgAPP/PS1 mice. Surprisingly, LPS injection decreased CD45 immunoreactivity in TgAPP/PS1 mice but increased it in non-Tg mice at 12 months. Although microglia in 12-month-old non-Tg mice showed stronger response to LPS than 2-month-old non-Tg mice, microglia in TgAPP/PS1 mice exhibited diminished immune response to LPS during aging. Our data indicate that microglial TLR4 signaling is altered in an AD mouse model and suggest that altered TLR4 signaling may contribute to Aß accumulation in the brain.


Subject(s)
Aging , Alzheimer Disease/genetics , Lipopolysaccharides/chemistry , Microglia/metabolism , Toll-Like Receptor 4/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Hippocampus/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Monocytes/metabolism , Signal Transduction
4.
Brain Behav Immun ; 53: 84-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26562537

ABSTRACT

Interleukin-17A (IL-17A) is generally considered as one of the pathogenic factors involved in multiple sclerosis (MS). Indirect evidence for this is that IL-17A-producing T helper 17 (Th17) cells preferentially accumulate in lesions of MS and experimental autoimmune encephalomyelitis (EAE). However, a direct involvement of IL-17A in MS pathogenesis is still an open question. In this study, we overexpressed IL-17A in the brains of mice (IL-17A-in-Brain mice) via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated gene delivery. In spite of high levels of IL-17A expression in the brain and blood, IL-17A-in-Brain mice exhibit no inflammatory responses and no abnormalities in motor coordination and spatial orientation. Unexpectedly, IL-17A-in-Brain mice show decreases in body weight and adipose tissue mass and an improvement in glucose tolerance and insulin sensitivity. IL-17A enhances glucose uptake in PC12 cells by activation of AKT. Our results provide direct evidence for the first time that IL-17A overexpression in the central nervous system does not cause physical and learning disabilities and neuroinflammation and suggest that IL-17A may regulate glucose metabolism through the AKT signaling pathway.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Glucose/metabolism , Interleukin-17/administration & dosage , Learning Disabilities/etiology , Proto-Oncogene Proteins c-akt/metabolism , Adipose Tissue/pathology , Animals , Cytokines/metabolism , Dependovirus/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Transfer Techniques , Insulin Resistance , Interleukin-17/biosynthesis , Interleukin-17/genetics , Learning Disabilities/metabolism , Learning Disabilities/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , PC12 Cells , Rats , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism
5.
Pharmaceutics ; 15(3)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36986581

ABSTRACT

The availability of several bioorthogonal reactions that can proceed selectively and efficiently under physiologically relevant conditions has garnered the interest of biochemists and organic chemists alike. Bioorthogonal cleavage reactions represent the latest innovation in click chemistry. Here, we employed the Staudinger ligation reaction to release radioactivity from immunoconjugates, improving target-to-background ratios. In this proof-of-concept study, model systems, including the anti-HER2 antibody trastuzumab, radioisotope I-131, and a newly synthesized bifunctional phosphine, were used. Staudinger ligation occurred when biocompatible N-glycosyl azides reacted with this radiolabeled immunoconjugate, leading to cleavage of the radioactive label from the molecule. We demonstrated this click cleavage in vitro and in vivo. Biodistribution studies in tumor models showed that radioactivity was eliminated from the bloodstream, thereby improving tumor-to-blood ratios. SPECT imaging revealed that tumors could be visualized with enhanced clarity. Our simple approach represents a novel application of bioorthogonal click chemistry in the development of antibody-based theranostics.

6.
Am J Pathol ; 179(3): 1095-103, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21763676

ABSTRACT

The accumulation of ß-amyloid protein (Aß) in the brain is thought to be a primary etiologic event in Alzheimer's disease (AD). Fibrillar Aß plaques, a hallmark of AD abnormality, are closely associated with activated microglia. Activated microglia have contradictory roles in the pathogenesis of AD, being either neuroprotective (by clearing harmful Aß and repairing damaged tissues) or neurotoxic (by producing proinflammatory cytokines and reactive oxygen species). Aß aggregates can activate microglia by interacting with multiple toll-like receptors (TLRs), the pattern-recognition receptors of the innate immune system. Because the adapter protein MyD88 is essential for the downstream signaling of all TLRs, except TLR3, we investigated the effects of MyD88 deficiency (MyD88(-/-)) on Aß accumulation and microglial activation in an AD mouse model. MyD88 deficiency decreased Aß load and microglial activation in the brain. The decrease in Aß load in an MyD88(-/-) AD mouse model was associated with increased and decreased protein expression of apolipoprotein E (apoE) and CX3CR1, respectively, compared with that in an MyD88 wild-type AD mouse model. These results suggest that MyD88 deficiency may reduce Aß load by enhancing the phagocytic capability of microglia through fractalkine (the ligand of CX3CR1) signaling and by promoting apoE-mediated clearance of Aß from the brain. These findings also suggest that chronic inflammatory responses induced by Aß accumulation via the MyD88-dependent signaling pathway exacerbate ß-amyloidosis in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/metabolism , Microglia/metabolism , Myeloid Differentiation Factor 88/deficiency , Animals , Apolipoproteins E/metabolism , CX3C Chemokine Receptor 1 , Cerebrum/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Inbred C57BL , Receptors, Chemokine/metabolism
7.
Sci Rep ; 12(1): 13360, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35922534

ABSTRACT

While boron neutron capture therapy (BNCT) depends primarily on the short flight range of the alpha particles emitted by the boron neutron capture reaction, gadolinium neutron capture therapy (GdNCT) mainly relies on gamma rays and Auger electrons released by the gadolinium neutron capture reaction. BNCT and GdNCT can be complementary in tumor therapy. Here, we studied the combined effects of BNCT and GdNCT when boron and gadolinium compounds were co-injected, followed by thermal neutron irradiation, and compared these effects with those of the single therapies. In cytotoxicity studies, some additive effects (32‒43%) were observed when CT26 cells were treated with both boron- and gadolinium-encapsulated PEGylated liposomes (B- and Gd-liposomes) compared to the single treatments. The tumor-suppressive effect was greater when BNCT was followed by GdNCT at an interval of 10 days rather than vice versa. However, tumor suppression with co-injection of B- and Gd-liposomes into tumor-bearing mice followed by neutron beam irradiation was comparable to that observed with Gd-liposome-only treatment but lower than B-liposome-only injection. No additive effect was observed with the combination of BNCT and GdNCT, which could be due to the shielding effect of gadolinium against thermal neutrons because of its overwhelmingly large thermal neutron cross section.


Subject(s)
Neoplasms , Neutron Capture Therapy , Animals , Boron , Boron Compounds , Disease Models, Animal , Gadolinium , Liposomes , Mice
8.
Pharmaceutics ; 14(8)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36015303

ABSTRACT

Triple-negative breast cancer (TNBC) does not express estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Because TNBC lacks the expression of commonly targeted receptors, it is challenging to develop a new imaging agent for this cancer subtype. Heterogeneous nuclear ribonucleoproteins (hnRNPs) are RNA-protein complexes that have been linked to tumor development and progression. Considering the high expression of hnRNPA2B1, an hnRNP subtype, in TNBC MDA-MB-231 cells, this study aimed to develop a novel hnRNPA2B1 antibody-based nuclear imaging agent. The hnRNPA2B1-specific antibody was radiolabeled with 64Cu and evaluated in vitro and in vivo. The trans-cyclooctene (TCO) was functionalized on the antibody to obtain hnRNP-PEG4-TCO and reactive tetrazine (Tz) on the ultrastable bifunctional chelator PCB-TE2A-alkyne to yield PCB-TE2A-Tz for the inverse electron demand Diels-Alder reaction. The 64Cu-radiolabeled antibody was administered and imaged at 1-18 h time points for conventional imaging. Alternatively, the unlabeled antibody conjugate was administered, and 48 h later radiolabeled 64Cu-PCB-TE2A-Tz was administered to the same mice for the pretargeting strategy and imaged at the same time intervals for direct comparison. The tumor was successfully visualized in both strategies, and comparatively, pretargeting showed superior results. The 64Cu-PCB-TE2A-Tz was successfully clicked at the tumor site with hnRNP-PEG4-TCO and the non-clicked were concurrently eliminated. This led to increase the tumor uptake with extremely high tumor-to-background ratio manifested by positron emission tomography (PET) imaging and biodistribution studies.

9.
J Neuroinflammation ; 8: 92, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21827663

ABSTRACT

BACKGROUND: Amyloid plaques, a pathological hallmark of Alzheimer's disease (AD), are accompanied by activated microglia. The role of activated microglia in the pathogenesis of AD remains controversial: either clearing Aß deposits by phagocytosis or releasing proinflammatory cytokines and cytotoxic substances. Microglia can be activated via toll-like receptors (TLRs), a class of pattern-recognition receptors in the innate immune system. We previously demonstrated that an AD mouse model homozygous for a loss-of-function mutation of TLR4 had increases in Aß deposits and buffer-soluble Aß in the brain as compared with a TLR4 wild-type AD mouse model at 14-16 months of age. However, it is unknown if TLR4 signaling is involved in initiation of Aß deposition as well as activation and recruitment of microglia at the early stage of AD. Here, we investigated the role of TLR4 signaling and microglial activation in early stages using 5-month-old AD mouse models when Aß deposits start. METHODS: Microglial activation and amyloid deposition in the brain were determined by immunohistochemistry in the AD models. Levels of cerebral soluble Aß were determined by ELISA. mRNA levels of cytokines and chemokines in the brain and Aß-stimulated monocytes were quantified by real-time PCR. Cognitive functions were assessed by the Morris water maze. RESULTS: While no difference was found in cerebral Aß load between AD mouse models at 5 months with and without TLR4 mutation, microglial activation in a TLR4 mutant AD model (TLR4M Tg) was less than that in a TLR4 wild-type AD model (TLR4W Tg). At 9 months, TLR4M Tg mice had increased Aß deposition and soluble Aß42 in the brain, which were associated with decrements in cognitive functions and expression levels of IL-1ß, CCL3, and CCL4 in the hippocampus compared to TLR4W Tg mice. TLR4 mutation diminished Aß-induced IL-1ß, CCL3, and CCL4 expression in monocytes. CONCLUSION: This is the first demonstration of TLR4-dependent activation of microglia at the early stage of ß-amyloidosis. Our results indicate that TLR4 is not involved in the initiation of Aß deposition and that, as Aß deposits start, microglia are activated via TLR4 signaling to reduce Aß deposits and preserve cognitive functions from Aß-mediated neurotoxicity.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Cognition Disorders/physiopathology , Microglia/physiology , Toll-Like Receptor 4/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/physiology , Brain/anatomy & histology , Brain/physiology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Humans , Maze Learning , Mice , Mice, Transgenic , Microglia/cytology , Mutation , Signal Transduction , Toll-Like Receptor 4/metabolism
10.
J Mol Neurosci ; 71(6): 1245-1255, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33185814

ABSTRACT

Repeated exposure to toll-like receptor 4 (TLR4) ligands, such as lipopolysaccharide (LPS), reduces responses of monocytes/macrophages to LPS (LPS/endotoxin tolerance). Microglial exposure to Aß deposits, a TLR4 ligand, may cause "Aß/LPS tolerance," leading to decreased Aß clearance. We demonstrated that microglial activation by LPS is diminished in Aß deposit-bearing 12-month-old model mice of Alzheimer's disease (AD), compared with non-AD mice and Aß deposit-free 2-month-old AD mice. Because miR-146a plays a predominant role in inducing TLR tolerance in macrophages and because miR-146a in extracellular vesicles (EVs) shed by inflammatory macrophages increases in circulation, we investigated potential roles of miR-146a and inflammatory EVs in inducing TLR tolerance in microglia and in altering expression of inflammatory AD risk genes. We found that miR-146a upregulation induces TLR tolerance and alters expression of inflammatory AD risk genes in response to LPS treatment in BV2 microglia. LPS brain injection altered expression of the AD risk genes in 12-month-old AD mice but not in non-AD littermates. EVs from inflammatory macrophages polarize BV2 microglia to M1 phenotype and induce TLR tolerance. Microglia exposed to Aß in the brain show reduced cytokine responses to systemic inflammation due to peripheral LPS injection, indicating TLR/Aß tolerance in microglia. Our results suggest that increased miR-146a induces microglial Aß/LPS tolerance and that circulating EVs shed by inflammatory macrophages contribute to microglial Aß/LPS tolerance, leading to reduced Aß clearance. Our study also suggests that altered expression of inflammatory AD risk genes may contribute to AD development via the same molecular mechanism underlying LPS tolerance.


Subject(s)
Alzheimer Disease/genetics , Exosomes/genetics , MicroRNAs/metabolism , Toll-Like Receptor 4/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Exosomes/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Microglia/drug effects , Microglia/metabolism , Presenilin-1/genetics , RAW 264.7 Cells , Signal Transduction
11.
Mol Neurobiol ; 51(1): 43-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24733587

ABSTRACT

Accumulation of amyloid beta-peptide (Aß) in the brain is hypothesized to be a causal event leading to dementia in Alzheimer's disease (AD). Aß vaccination removes Aß deposits from the brain. Aß immunotherapy, however, may cause T cell- and/or Fc-receptor-mediated brain inflammation and relocate parenchymal Aß deposits to blood vessels leading to cerebral hemorrhages. Because catalytic antibodies do not form stable immune complexes and Aß fragments produced by catalytic antibodies are less likely to form aggregates, Aß-specific catalytic antibodies may have safer therapeutic profiles than reversibly-binding anti-Aß antibodies. Additionally, catalytic antibodies may remove Aß more efficiently than binding antibodies because a single catalytic antibody can hydrolyze thousands of Aß molecules. We previously isolated Aß-specific catalytic antibody, IgVL5D3, with strong Aß-hydrolyzing activity. Here, we evaluated the prophylactic and therapeutic efficacy of brain-targeted IgVL5D3 gene delivery via recombinant adeno-associated virus serotype 9 (rAAV9) in an AD mouse model. One single injection of rAAV9-IgVL5D3 into the right ventricle of AD model mice yielded widespread, high expression of IgVL5D3 in the unilateral hemisphere. IgVL5D3 expression was readily detectable in the contralateral hemisphere but to a much lesser extent. IgVL5D3 expression was also confirmed in the cerebrospinal fluid. Prophylactic and therapeutic injection of rAAV9-IgVL5D3 reduced Aß load in the ipsilateral hippocampus of AD model mice. No evidence of hemorrhages, increased vascular amyloid deposits, increased proinflammatory cytokines, or infiltrating T-cells in the brains was found in the experimental animals. AAV9-mediated anti-Aß catalytic antibody brain delivery can be prophylactic and therapeutic options for AD.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Disease/therapy , Biocatalysis , Genes, Immunoglobulin , Genetic Therapy , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Animals , Antibodies/pharmacology , Biocatalysis/drug effects , Cerebral Amyloid Angiopathy/etiology , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Dependovirus/metabolism , Disease Models, Animal , Genetic Vectors/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation/pathology , Injections , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Plaque, Amyloid/metabolism , Solubility
12.
J Neuroimmunol ; 244(1-2): 70-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22326143

ABSTRACT

We evaluated the therapeutic efficacy of combined treatment of Aß-immunization with simvastatin in an Alzheimer mouse model at age 22 months. DNA prime-adenovirus boost immunization induced modest anti-Aß titers and simvastatin increased the seropositive rate. Aß-KLH was additionally administered to boost the titers. Irrespective of simvastatin, the immunization did not decrease cerebral Aß deposits but increased soluble Aß and tended to exacerbate amyloid angiopathy in the hippocampus. The immunization increased cerebral invasion of leukocytes and simvastatin counteracted the increase. Thus, modest anti-Aß titers can increase soluble Aß and simvastatin may reduce inflammation associated with vaccination in aged Alzheimer mouse models.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Simvastatin/therapeutic use , Alzheimer Disease/immunology , Alzheimer Disease/prevention & control , Alzheimer Vaccines/therapeutic use , Animals , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Amyloid Angiopathy/immunology , Cerebral Amyloid Angiopathy/prevention & control , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Behav Brain Res ; 227(1): 36-42, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22051943

ABSTRACT

Toll-like receptors (TLRs) are a family of pattern-recognition receptors in innate immunity and provide a first line defense against pathogens and tissue injuries. In addition to important roles in infection, inflammation, and immune diseases, recent studies show that TLR signaling is involved in modulation of learning, memory, mood, and neurogenesis. Because MyD88 is essential for the downstream signaling of all TLRs, except TLR3, we investigated the effects of MyD88 deficiency (MyD88-/-) on behavioral functions in mice. Additionally, we recently demonstrated that a mouse model of Alzheimer's disease (AD) deficient for MyD88 had decreases in Aß deposits and soluble Aß in the brain as compared with MyD88 sufficient AD mouse models. Because accumulation of Aß in the brain is postulated to be a causal event leading to cognitive deficits in AD, we investigated the effects of MyD88 deficiency on behavioral functions in the AD mouse model at 10 months of age. MyD88 deficient mice showed more anxiety in the elevated plus-maze. In the motor coordination tests, MyD88 deficient mice remained on a beam and a bar for a longer time, but with slower initial movement on the bar. In the Morris water maze test, MyD88 deficiency appeared to improve spatial learning irrespective of the transgene. Our findings suggest that the MyD88-dependent pathway contributes to behavioral functions in an AD mouse model and its control group.


Subject(s)
Alzheimer Disease/complications , Anxiety/etiology , Exploratory Behavior/physiology , Motor Activity/physiology , Myeloid Differentiation Factor 88/deficiency , Psychomotor Performance/physiology , Space Perception/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Humans , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Time Factors
14.
J Alzheimers Dis ; 27(1): 23-38, 2011.
Article in English | MEDLINE | ID: mdl-21709371

ABSTRACT

Accumulation of amyloid-ß protein (Aß) in the brain is thought to be a causal event in Alzheimer's disease (AD). Immunotherapy targeting Aß holds great promise for reducing Aß in the brain. Here, we evaluated the efficacy and safety of anti-Aß single-chain antibody (scFv59) delivery via recombinant adeno-associated virus (rAAV) on reducing Aß deposits in an AD mouse model (TgAßPPswe/PS1dE9). First, delivery of scFv59 to the brain was optimized by injecting rAAV serotypes 1, 2, and 5 into the right lateral ventricle. Symmetrical high expression of scFv59 was found throughout the hippocampus and partly in the neocortex in both hemispheres via rAAV1 or rAAV5, while scFv59 expression via rAAV2 was mostly limited to one hemisphere. rAAV1, however, induced apoptosis and microglial activation but rAAV5 did not. Therefore, rAAV5 was selected for therapeutic scFv59 delivery in TgAßPPswe/PS1dE9 mice. rAAV5 was similarly injected into the ventricle of 10-month-old TgAßPPswe/PS1dE9 mice and 5 months later its efficacy and safety were evaluated. Immunoreactive Aß deposits reduced in the hippocampus. Aß42 levels in cerebrospinal fluid (CSF) tended to increase and the Aß40 : 42 ratio decreased in CSF, suggesting that Aß42 was relocated from the parenchyma to CSF. Hemorrhages associated with a focal increase in blood vessel amyloid were found in the brain. While immunotherapy has great potential for clearing cerebral Aß, caution for cerebrovascular effects should be exercised when rAAV-mediated anti-Aß immunotherapy is applied.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Amyloidogenic Proteins/metabolism , Cerebral Hemorrhage/chemically induced , Single-Chain Antibodies/adverse effects , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor , Analysis of Variance , Animals , Antigens, CD , Apoptosis/drug effects , Apoptosis/genetics , Brain/metabolism , Brain/pathology , Cell Line, Transformed , Dependovirus/physiology , Disease Models, Animal , Drug Delivery Systems , Enzyme-Linked Immunosorbent Assay/methods , Glial Fibrillary Acidic Protein/metabolism , Immunotherapy , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1 , Transduction, Genetic , Transfection
15.
Pharmacogenet Genomics ; 18(9): 781-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18698231

ABSTRACT

BACKGROUND: Genetic variation in mRNA expression plays a critical role in human phenotypic diversity, but it has proven difficult to detect regulatory polymorphisms - mostly single nucleotide polymorphisms (rSNPs). Additionally, variants in the transcribed region, termed here 'structural RNA SNPs' (srSNPs), can affect mRNA processing and turnover. Both rSNPs and srSNPs cause allelic mRNA expression imbalance (AEI) in heterozygous individuals. We have used AEI to discover and characterize regulatory polymorphisms in OPRM1, TPH2, MDR1, DRD2, and VKORC1. The objective of this study was to use AEI to determine the extent of cis-regulatory factors in pharmacogenetic genes. METHODS: We applied a rapid and accurate AEI methodology for testing 42 genes implicated in cardiovascular and central nervous system diseases, and affecting drug metabolism and transport. Each gene was analyzed in physiologically relevant human autopsy tissues, including brain, heart, liver, intestines, and lymphocytes. RESULTS: Substantial AEI was observed in approximately 55% of the surveyed genes. Focusing on cardiovascular candidate genes in human hearts, AEI analysis revealed frequent cis-acting regulatory factors in ACE and SOD2 mRNA expression, having potential clinical significance. SNP scanning to locate regulatory polymorphisms in a number of genes failed to support several previously proposed promoter SNPs discovered with use of reporter gene assays in heterologous tissues, while srSNPs appear more frequent than expected. Computational analysis of mRNA folding indicates that approximately 90% of srSNPs affect mRNA folding, and hence potentially function. CONCLUSION: Our results indicate that both rSNPs and srSNPs represent a still largely untapped reservoir of variants that contribute to human phenotypic diversity.


Subject(s)
Allelic Imbalance , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Transcription, Genetic , Autopsy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Pharmacogenetics , Polymerase Chain Reaction , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL