Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Database
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(32): e2306731120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37523555

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal disease affecting upper and lower motor neurons. Microglia directly interact with motor neurons and participate in the progression of ALS. Single-cell mass cytometry (CyTOF) analysis revealed prominent expression of α5 integrin in microglia and macrophages in a superoxide dismutase-1 G93A mouse model of ALS (SOD1G93A). In postmortem tissues from ALS patients with various clinical ALS phenotypes and disease duration, α5 integrin is prominent in motor pathways of the central and peripheral nervous system and in perivascular zones associated with the blood-brain barrier. In SOD1G93A mice, administration of a monoclonal antibody against α5 integrin increased survival compared to an isotype control and improved motor function on behavioral testing. Together, these findings in mice and in humans suggest that α5 integrin is a potential therapeutic target in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Cortex , Mice , Humans , Animals , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Integrin alpha5/metabolism , Mice, Transgenic , Superoxide Dismutase/metabolism , Macrophages/metabolism , Disease Models, Animal
2.
Nat Commun ; 13(1): 5451, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36114178

ABSTRACT

Alzheimer's disease (AD) is the consequence of neuronal death and brain atrophy associated with the aggregation of protein tau into fibrils. Thus disaggregation of tau fibrils could be a therapeutic approach to AD. The small molecule EGCG, abundant in green tea, has long been known to disaggregate tau and other amyloid fibrils, but EGCG has poor drug-like properties, failing to fully penetrate the brain. Here we have cryogenically trapped an intermediate of brain-extracted tau fibrils on the kinetic pathway to EGCG-induced disaggregation and have determined its cryoEM structure. The structure reveals that EGCG molecules stack in polar clefts between the paired helical protofilaments that pathologically define AD. Treating the EGCG binding position as a pharmacophore, we computationally screened thousands of drug-like compounds for compatibility for the pharmacophore, discovering several that experimentally disaggregate brain-derived tau fibrils in vitro. This work suggests the potential of structure-based, small-molecule drug discovery for amyloid diseases.


Subject(s)
Alzheimer Disease , Amyloidosis , tau Proteins , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid/drug effects , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Cryoelectron Microscopy , Drug Evaluation, Preclinical/methods , Humans , Tea/chemistry , tau Proteins/chemistry , tau Proteins/drug effects , tau Proteins/metabolism
3.
J Exp Med ; 216(3): 539-555, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30770411

ABSTRACT

It has been challenging to produce ex vivo models of the inclusion pathologies that are hallmark pathologies of many neurodegenerative diseases. Using three-dimensional mouse brain slice cultures (BSCs), we have developed a paradigm that rapidly and robustly recapitulates mature neurofibrillary inclusion and Lewy body formation found in Alzheimer's and Parkinson's disease, respectively. This was achieved by transducing the BSCs with recombinant adeno-associated viruses (rAAVs) that express α-synuclein or variants of tau. Notably, the tauopathy BSC model enables screening of small molecule therapeutics and tracking of neurodegeneration. More generally, the rAAV BSC "toolkit" enables efficient transduction and transgene expression from neurons, microglia, astrocytes, and oligodendrocytes, alone or in combination, with transgene expression lasting for many months. These rAAV-based BSC models provide a cost-effective and facile alternative to in vivo studies, and in the future can become a widely adopted methodology to explore physiological and pathological mechanisms related to brain function and dysfunction.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Dependovirus/genetics , Parkinson Disease/pathology , Alzheimer Disease/virology , Animals , Brain/metabolism , Brain/virology , Drug Evaluation, Preclinical/methods , Gene Expression , Humans , Mice, Inbred C3H , Mice, Transgenic , Microorganisms, Genetically-Modified , Mutation , Neurons/pathology , Organ Culture Techniques , Parkinson Disease/virology , Transduction, Genetic , Transgenes , alpha-Synuclein/genetics , tau Proteins/genetics
4.
Nat Med ; 20(12): 1444-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401692

ABSTRACT

Neurodegenerative diseases, such as frontotemporal dementia (FTD), are often associated with behavioral deficits, but the underlying anatomical and molecular causes remain poorly understood. Here we show that forebrain-specific expression of FTD-associated mutant CHMP2B in mice causes several age-dependent neurodegenerative phenotypes, including social behavioral impairments. The social deficits were accompanied by a change in AMPA receptor (AMPAR) composition, leading to an imbalance between Ca(2+)-permeable and Ca(2+)-impermeable AMPARs. Expression of most AMPAR subunits was regulated by the brain-enriched microRNA miR-124, whose abundance was markedly decreased in the superficial layers of the cerebral cortex of mice expressing the mutant CHMP2B. We found similar changes in miR-124 and AMPAR levels in the frontal cortex and induced pluripotent stem cell-derived neurons from subjects with behavioral variant FTD. Moreover, ectopic miR-124 expression in the medial prefrontal cortex of mutant mice decreased AMPAR levels and partially rescued behavioral deficits. Knockdown of the AMPAR subunit Gria2 also alleviated social impairments. Our results identify a previously undescribed mechanism involving miR-124 and AMPARs in regulating social behavior in FTD and suggest a potential therapeutic avenue.


Subject(s)
Behavior, Animal , Endosomal Sorting Complexes Required for Transport/genetics , Frontal Lobe/metabolism , Frontotemporal Dementia/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, AMPA/metabolism , Social Behavior , Animals , Calcium/metabolism , Cerebral Cortex/metabolism , Disease Models, Animal , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/psychology , Mice , Mice, Transgenic , Prefrontal Cortex/metabolism
5.
Rinsho Shinkeigaku ; 51(8): 603-7, 2011 Aug.
Article in Japanese | MEDLINE | ID: mdl-21878728

ABSTRACT

We investigated fixed basal ganglia specimens, including globus pallidus and putamen, with 21.1-Tesla MRI allowing us to achieve a microscopic level resolution from a patient with pathologically confirmed dementia with Lewy bodies (DLB) and a neurologically normal control case. We acquired T2 and T2 * weighted images that demonstrated diffuse and patchy lower intensities in the basal ganglia compared to control. There are several paramagnetic substances in brain tissue that could potentially reduce both T2 and T2 * relaxation times, including ferritin, iron (Fe3+), manganese, copper and others. Because iron is most abundant, low intensities on T2 and T2 * weighted images most likely reflect iron deposition. Iron, especially Fe3+, deposition was visible in the pathological specimens stained with Prussian blue after images were obtained. Although radiological-pathological comparisons are not straightforward with respect to either the MRI signal or relaxation quantification, there appears to be a correlation between the relative increase in iron as assessed by Prussian blue staining and the decrease in T2 * value between the DLB and control specimens. As such, this exceptionally high field MRI technique may provide details about the role that iron deposition plays either directly or indirectly as a biomarker in neurodegenerative processes.


Subject(s)
Lewy Body Disease/pathology , Magnetic Field Therapy/methods , Aged, 80 and over , Basal Ganglia/pathology , Humans , Iron/analysis , Male
6.
J Neurosci ; 28(13): 3277-90, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18367595

ABSTRACT

Huntington's disease (HD) is a hereditary neurological disease caused by expended CAG repeats in the HD gene, which codes for a protein called Huntingtin (Htt). The resultant mutant Huntingtin (mHtt) forms aggregates in neurons and causes neuronal dysfunction. In astrocytes, the largest population of brain cells, mHtt also exists. We report herein that astrocyte-conditioned medium (ACM) collected from astrocytes of R6/2 mice (a mouse model of HD) caused primary cortical neurons to grow less-mature neurites, migrate more slowly, and exhibit lower calcium influx after depolarization than those maintained in wild-type (WT) ACM. Using a cytokine antibody array and ELISA assays, we demonstrated that the amount of a chemokine [chemokine (C-C motif) ligand 5 (CCL5)/regulated on activation normal T cell expressed and secreted (RANTES)] released by R6/2 astrocytes was much less than that by WT astrocytes. When cortical neurons were treated with the indicated ACM, supplementation with recombinant CCL5/RANTES ameliorated the neuronal deficiency caused by HD-ACM, whereas removing CCL5/RANTES from WT-ACM using an anti-CCL5/RANTES antibody mimicked the effects evoked by HD-ACM. Quantitative PCR and promoter analyses demonstrated that mHtt hindered the activation of the CCL5/RANTES promoter by reducing the availability of nuclear factor kappaB-p65 and, hence, reduced the transcript level of CCL5/RANTES. Moreover, ELISA assays and immunocytochemical staining revealed that mHtt retained the residual CCL5/RANTES inside R6/2 astrocytes. In line with the above findings, elevated cytosolic CCL5/RANTES levels were also observed in the brains of two mouse models of HD [R6/2 and Hdh((CAG)150)] and human HD patients. These findings suggest that mHtt hinders one major trophic function of astrocytes which might contribute to the neuronal dysfunction of HD.


Subject(s)
Astrocytes/metabolism , Chemokine CCL5/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Nuclear Proteins/physiology , Trinucleotide Repeat Expansion/physiology , Aged , Aged, 80 and over , Animals , Animals, Newborn , Astrocytes/chemistry , Brain/pathology , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Chromatin Immunoprecipitation/methods , Culture Media, Conditioned/pharmacology , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Proteins/genetics , Rats , Rats, Sprague-Dawley , Transfection/methods , Trinucleotide Repeat Expansion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL