Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 172(5): 952-965.e18, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474921

ABSTRACT

Viruses that are typically benign sometimes invade the brainstem in otherwise healthy children. We report bi-allelic DBR1 mutations in unrelated patients from different ethnicities, each of whom had brainstem infection due to herpes simplex virus 1 (HSV1), influenza virus, or norovirus. DBR1 encodes the only known RNA lariat debranching enzyme. We show that DBR1 expression is ubiquitous, but strongest in the spinal cord and brainstem. We also show that all DBR1 mutant alleles are severely hypomorphic, in terms of expression and function. The fibroblasts of DBR1-mutated patients contain higher RNA lariat levels than control cells, this difference becoming even more marked during HSV1 infection. Finally, we show that the patients' fibroblasts are highly susceptible to HSV1. RNA lariat accumulation and viral susceptibility are rescued by wild-type DBR1. Autosomal recessive, partial DBR1 deficiency underlies viral infection of the brainstem in humans through the disruption of tissue-specific and cell-intrinsic immunity to viruses.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Brain Stem/metabolism , Brain Stem/virology , RNA/chemistry , RNA/metabolism , Alleles , Amino Acid Sequence , Animals , Brain Diseases, Metabolic, Inborn/pathology , Brain Stem/pathology , Encephalitis, Viral/genetics , Escherichia coli/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human , Humans , Interferons/metabolism , Introns/genetics , Male , Mice , Mutant Proteins/metabolism , Mutation/genetics , Open Reading Frames/genetics , Pedigree , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/deficiency , RNA Nucleotidyltransferases/genetics , Toll-Like Receptor 3/metabolism , Virus Replication
2.
J Neurosci ; 41(19): 4305-4320, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33888602

ABSTRACT

Vascular dysfunction is a universal feature of aging and decreased cerebral blood flow has been identified as an early event in the pathogenesis of Alzheimer's disease (AD). Cerebrovascular dysfunction in AD includes deficits in neurovascular coupling (NVC), a mechanism that ensures rapid delivery of energy substrates to active neurons through the blood supply. The mechanisms underlying NVC impairment in AD, however, are not well understood. We have previously shown that mechanistic/mammalian target of rapamycin (mTOR) drives cerebrovascular dysfunction in models of AD by reducing the activity of endothelial nitric oxide synthase (eNOS), and that attenuation of mTOR activity with rapamycin is sufficient to restore eNOS-dependent cerebrovascular function. Here we show mTOR drives NVC impairments in an AD model through the inhibition of neuronal NOS (nNOS)- and non-NOS-dependent components of NVC, and that mTOR attenuation with rapamycin is sufficient to restore NVC and even enhance it above WT responses. Restoration of NVC and concomitant reduction of cortical amyloid-ß levels effectively treated memory deficits in 12-month-old hAPP(J20) mice. These data indicate that mTOR is a critical driver of NVC dysfunction and underlies cognitive impairment in an AD model. Together with our previous findings, the present studies suggest that mTOR promotes cerebrovascular dysfunction in AD, which is associated with early disruption of nNOS activation, through its broad negative impact on nNOS as well as on non-NOS components of NVC. Our studies highlight the potential of mTOR attenuation as an efficacious treatment for AD and potentially other neurologic diseases of aging.SIGNIFICANCE STATEMENT Failure of the blood flow response to neuronal activation [neurovascular coupling (NVC)] in a model of AD precedes the onset of AD-like cognitive symptoms and is driven, to a large extent, by mammalian/mechanistic target of rapamycin (mTOR)-dependent inhibition of nitric oxide synthase activity. Our studies show that mTOR also drives AD-like failure of non-nitric oxide (NO)-mediated components of NVC. Thus, mTOR attenuation may serve to treat AD, where we find that neuronal NO synthase is profoundly reduced early in disease progression, and potentially other neurologic diseases of aging with cerebrovascular dysfunction as part of their etiology.


Subject(s)
Alzheimer Disease/drug therapy , Memory Disorders/drug therapy , Neurovascular Coupling/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebrovascular Disorders/physiopathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/psychology , Fear/psychology , Female , Humans , Male , Memory Disorders/psychology , Mice , Mice, Transgenic , Microvessels/pathology , Microvessels/ultrastructure , Nitric Oxide Synthase Type III/metabolism , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/genetics
3.
Am J Physiol Heart Circ Physiol ; 314(4): H693-H703, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351469

ABSTRACT

An intact blood-brain barrier (BBB) limits entry of proinflammatory and neurotoxic blood-derived factors into the brain parenchyma. The BBB is damaged in Alzheimer's disease (AD), which contributes significantly to the progression of AD pathologies and cognitive decline. However, the mechanisms underlying BBB breakdown in AD remain elusive, and no interventions are available for treatment or prevention. We and others recently established that inhibition of the mammalian/mechanistic target of rapamycin (mTOR) pathway with rapamycin yields significant neuroprotective effects, improving cerebrovascular and cognitive function in mouse models of AD. To test whether mTOR inhibition protects the BBB in neurological diseases of aging, we treated hAPP(J20) mice modeling AD and low-density lipoprotein receptor-null (LDLR-/-) mice modeling vascular cognitive impairment with rapamycin. We found that inhibition of mTOR abrogates BBB breakdown in hAPP(J20) and LDLR-/- mice. Experiments using an in vitro BBB model indicated that mTOR attenuation preserves BBB integrity through upregulation of specific tight junction proteins and downregulation of matrix metalloproteinase-9 activity. Together, our data establish mTOR activity as a critical mediator of BBB breakdown in AD and, potentially, vascular cognitive impairment and suggest that rapamycin and/or rapalogs could be used for the restoration of BBB integrity. NEW & NOTEWORTHY This report establishes mammalian/mechanistic target of rapamycin as a critical mediator of blood-brain barrier breakdown in models of Alzheimer's disease and vascular cognitive impairment and suggests that drugs targeting the target of rapamycin pathway could be used for the restoration of blood-brain barrier integrity in disease states.


Subject(s)
Alzheimer Disease/drug therapy , Behavior, Animal , Blood-Brain Barrier/drug effects , Cognition , Dementia, Vascular/drug therapy , Protein Kinase Inhibitors/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Cell Line , Dementia, Vascular/enzymology , Dementia, Vascular/pathology , Dementia, Vascular/psychology , Disease Models, Animal , Female , Male , Matrix Metalloproteinase 9/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , TOR Serine-Threonine Kinases/metabolism , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/enzymology , Tight Junctions/pathology
4.
J Neuroinflammation ; 11: 143, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25116321

ABSTRACT

BACKGROUND: Immune system cells are known to affect loss of neurons due to injury or disease. Recruitment of immune cells following retinal/CNS injury has been shown to affect the health and survival of neurons in several models. We detected close, physical contact between dendritic cells and retinal ganglion cells following an optic nerve crush, and sought to understand the underlying mechanisms. METHODS: CD11c-DTR/GFP mice producing a chimeric protein of diphtheria toxin receptor (DTR) and GFP from a transgenic CD11c promoter were used in conjunction with mice deficient in MyD88 and/or TRIF. Retinal ganglion cell injury was induced by an optic nerve crush, and the resulting interactions of the GFPhi cells and retinal ganglion cells were examined. RESULTS: Recruitment of GFPhi dendritic cells to the retina was significantly compromised in MyD88 and TRIF knockout mice. GFPhi dendritic cells played a significant role in clearing fluorescent-labeled retinal ganglion cells post-injury in the CD11c-DTR/GFP mice. In the TRIF and MyD88 deficient mice, the resting level of GFPhi dendritic cells was lower, and their influx was reduced following the optic nerve crush injury. The reduction in GFPhi dendritic cell numbers led to their replacement in the uptake of fluorescent-labeled debris by GFPlo microglia/macrophages. Depletion of GFPhi dendritic cells by treatment with diphtheria toxin also led to their displacement by GFPlo microglia/macrophages, which then assumed close contact with the injured neurons. CONCLUSIONS: The contribution of recruited cells to the injury response was substantial, and regulated by MyD88 and TRIF. However, the presence of these adaptor proteins was not required for interaction with neurons, or the phagocytosis of debris. The data suggested a two-niche model in which resident microglia were maintained at a constant level post-optic nerve crush, while the injury-stimulated recruitment of dendritic cells and macrophages led to their transient appearance in numbers equivalent to or greater than the resident microglia.


Subject(s)
Adaptor Proteins, Vesicular Transport/deficiency , Cell Movement/genetics , Dendritic Cells/physiology , Myeloid Differentiation Factor 88/deficiency , Retinal Ganglion Cells/pathology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antigens, Differentiation/metabolism , CD11c Antigen/genetics , CD11c Antigen/metabolism , Dendritic Cells/drug effects , Diphtheria Toxin/pharmacology , Disease Models, Animal , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Myeloid Cells/physiology , Myeloid Differentiation Factor 88/genetics , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Time Factors , Visual Pathways/pathology
5.
J Neurochem ; 124(6): 880-93, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23121022

ABSTRACT

Rapamycin, an inhibitor of target-of-rapamycin, extends lifespan in mice, possibly by delaying aging. We recently showed that rapamycin halts the progression of Alzheimer's (AD)-like deficits, reduces amyloid-beta (Aß) and induces autophagy in the human amyloid precursor protein (PDAPP) mouse model. To delineate the mechanisms by which chronic rapamycin delays AD we determined proteomic signatures in brains of control- and rapamycin-treated PDAPP mice. Proteins with reported chaperone-like activity were overrepresented among proteins up-regulated in rapamycin-fed PDAPP mice and the master regulator of the heat-shock response, heat-shock factor 1, was activated. This was accompanied by the up-regulation of classical chaperones/heat shock proteins (HSPs) in brains of rapamycin-fed PDAPP mice. The abundance of most HSP mRNAs except for alpha B-crystallin, however, was unchanged, and the cap-dependent translation inhibitor 4E-BP was active, suggesting that increased expression of HSPs and proteins with chaperone activity may result from preferential translation of pre-existing mRNAs as a consequence of inhibition of cap-dependent translation. The effects of rapamycin on the reduction of Aß, up-regulation of chaperones, and amelioration of AD-like cognitive deficits were recapitulated by transgenic over-expression of heat-shock factor 1 in PDAPP mice. These results suggest that, in addition to inducing autophagy, rapamycin preserves proteostasis by increasing chaperones. We propose that the failure of proteostasis associated with aging may be a key event enabling AD, and that chronic inhibition of target-of-rapamycin may delay AD by maintaining proteostasis in brain. Read the Editorial Highlight for this article on doi: 10.1111/jnc.12098.


Subject(s)
Alzheimer Disease/drug therapy , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Phenotype , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/biosynthesis , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Heat Shock Transcription Factors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/physiology , Up-Regulation/genetics
6.
Geroscience ; 45(3): 1987-1996, 2023 06.
Article in English | MEDLINE | ID: mdl-37052770

ABSTRACT

Peripheral artery disease (PAD), defined as reduced blood flow to the lower limbs, is a serious disorder that can lead to loss of function in the lower extremities and even loss of limbs. One of the main risk factors for PAD is age, with up to 25% of adults over the age of 55 and up to 40% over the age of 80 presenting with some form of the disease. While age is the largest risk factor for PAD, other risk factors include atherosclerosis, smoking, hypertension, and diabetes. Furthermore, previous studies have suggested that the incidence of PAD is significantly increased in patients with Alzheimer's disease (AD). Attenuation of mTOR with rapamycin significantly improves cerebral blood flow and heart function in aged rodents as well as in mouse models of atherosclerosis, atherosclerosis-driven cognitive impairment, and AD. In this study, we show that rapamycin treatment improves peripheral blood flow in aged mice and in mouse models of atherosclerosis and AD. Inhibition of mTOR with rapamycin ameliorates deficits in baseline hind paw perfusion in aged mice and restores levels of blood flow to levels indistinguishable from those of young controls. Furthermore, rapamycin treatment ameliorates peripheral blood flow deficits in mouse models of atherosclerosis and AD. These data indicate that mTOR is causally involved in the reduction of blood flow to lower limbs associated with aging, atherosclerosis, and AD-like progression in model mice. Rapamycin or other mTOR inhibitors may have potential as interventions to treat peripheral artery disease and other peripheral circulation-related conditions.


Subject(s)
Alzheimer Disease , Atherosclerosis , Peripheral Arterial Disease , Mice , Animals , Alzheimer Disease/complications , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Atherosclerosis/drug therapy , Peripheral Arterial Disease/drug therapy , Peripheral Arterial Disease/complications
7.
Nat Commun ; 14(1): 2367, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185259

ABSTRACT

Vascular mechanisms of Alzheimer's disease (AD) may constitute a therapeutically addressable biological pathway underlying dementia. We previously demonstrated that soluble pathogenic forms of tau (tau oligomers) accumulate in brain microvasculature of AD and other tauopathies, including prominently in microvascular endothelial cells. Here we show that soluble pathogenic tau accumulates in brain microvascular endothelial cells of P301S(PS19) mice modeling tauopathy and drives AD-like brain microvascular deficits. Microvascular impairments in P301S(PS19) mice were partially negated by selective removal of pathogenic soluble tau aggregates from brain. We found that similar to trans-neuronal transmission of pathogenic forms of tau, soluble tau aggregates are internalized by brain microvascular endothelial cells in a heparin-sensitive manner and induce microtubule destabilization, block endothelial nitric oxide synthase (eNOS) activation, and potently induce endothelial cell senescence that was recapitulated in vivo in microvasculature of P301S(PS19) mice. Our studies suggest that soluble pathogenic tau aggregates mediate AD-like brain microvascular deficits in a mouse model of tauopathy, which may arise from endothelial cell senescence and eNOS dysfunction triggered by internalization of soluble tau aggregates.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Animals , tau Proteins/genetics , tau Proteins/metabolism , Endothelial Cells/metabolism , Tauopathies/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Disease Models, Animal , Cellular Senescence , Mice, Transgenic
8.
Geroscience ; 43(1): 115-124, 2021 02.
Article in English | MEDLINE | ID: mdl-33063253

ABSTRACT

The ability to generate in vitro cultures of neuronal cells has been instrumental in advancing our understanding of the nervous system. Rodent models have been the principal source of brain cells used in primary cultures for over a century, providing insights that are widely applicable to human diseases. However, therapeutic agents that showed benefit in rodent models, particularly those pertaining to aging and age-associated dementias, have frequently failed in clinical trials. This discrepancy established a potential "translational gap" between human and rodent studies that may at least partially be explained by the phylogenetic distance between rodent and primate species. Several non-human primate (NHP) species, including the common marmoset (Callithrix jacchus), have been used extensively in neuroscience research, but in contrast to rodent models, practical approaches to the generation of primary cell culture systems amenable to molecular studies that can inform in vivo studies are lacking. Marmosets are a powerful model in biomedical research and particularly in studies of aging and age-associated diseases because they exhibit an aging phenotype similar to humans. Here, we report a practical method to culture primary marmoset neurons and astrocytes from brains of medically euthanized postnatal day 0 (P0) marmoset newborns that yield highly pure primary neuron and astrocyte cultures. Primary marmoset neuron and astrocyte cultures can be generated reliably to provide a powerful NHP in vitro model in neuroscience research that may enable mechanistic studies of nervous system aging and of age-related neurodegenerative disorders. Because neuron and astrocyte cultures can be used in combination with in vivo approaches in marmosets, primary marmoset neuron and astrocyte cultures may help bridge the current translational gap between basic and clinical studies in nervous system aging and age-associated neurological diseases.


Subject(s)
Astrocytes , Callithrix , Aging , Animals , Neurons , Phylogeny
9.
J Neurochem ; 113(6): 1481-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345760

ABSTRACT

Our previous work demonstrated that immunoproteasome is up-regulated in the retina and brain in response to injury that does not involve an inflammatory response (J. Neurochem. 2008; 106:158). These results suggest additional non-immune functions for the immunoproteasome in the cellular stress response pathway. The present study further investigates the potential involvement of the immunoproteasome in responding to the chronic stress of aging or oxidant exposure in the retina and cultured retinal pigment epithelial (RPE) cells from knock-out mice missing either one (lmp7(-/-)) or two (lmp7(-/-)/mecl-1(-/-)) immunoproteasome subunits. We show that aging and chronic oxidative stress up-regulates immunoproteasome in the retina and RPE from wild-type mice. No up-regulation of LMP2 was observed in retinas or RPE lacking MECL-1 and/or LMP7, suggesting that the full complement of immunoproteasome subunits is required to achieve maximal up-regulation in response to stress. We also show that RPE deficient in immunoproteasome are more susceptible to oxidation-induced cell death, supporting a role for immunoproteasome in protecting from oxidative stress. These results provide key mechanistic insight into novel aspects of proteasome biology and are an important first step in identifying alternative roles for retinal immunoproteasome that are unrelated to its role in the immune response.


Subject(s)
Cysteine Endopeptidases/deficiency , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Oxidative Stress/physiology , Receptors, Cytoplasmic and Nuclear/deficiency , Retinal Pigment Epithelium/cytology , Up-Regulation/physiology , Aging , Analysis of Variance , Animals , Cells, Cultured , Cysteine Endopeptidases/genetics , Epithelial Cells/drug effects , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/pharmacology , Proteasome Endopeptidase Complex , Protein Subunits/genetics , Protein Subunits/metabolism , Retinal Pigment Epithelium/drug effects , Up-Regulation/drug effects
10.
Aging Cell ; 19(1): e13057, 2020 01.
Article in English | MEDLINE | ID: mdl-31693798

ABSTRACT

Cerebrovascular dysfunction and cognitive decline are highly prevalent in aging, but the mechanisms underlying these impairments are unclear. Cerebral blood flow decreases with aging and is one of the earliest events in the pathogenesis of Alzheimer's disease (AD). We have previously shown that the mechanistic/mammalian target of rapamycin (mTOR) drives disease progression in mouse models of AD and in models of cognitive impairment associated with atherosclerosis, closely recapitulating vascular cognitive impairment. In the present studies, we sought to determine whether mTOR plays a role in cerebrovascular dysfunction and cognitive decline during normative aging in rats. Using behavioral tools and MRI-based functional imaging, together with biochemical and immunohistochemical approaches, we demonstrate that chronic mTOR attenuation with rapamycin ameliorates deficits in learning and memory, prevents neurovascular uncoupling, and restores cerebral perfusion in aged rats. Additionally, morphometric and biochemical analyses of hippocampus and cortex revealed that mTOR drives age-related declines in synaptic and vascular density during aging. These data indicate that in addition to mediating AD-like cognitive and cerebrovascular deficits in models of AD and atherosclerosis, mTOR drives cerebrovascular, neuronal, and cognitive deficits associated with normative aging. Thus, inhibitors of mTOR may have potential to treat age-related cerebrovascular dysfunction and cognitive decline. Since treatment of age-related cerebrovascular dysfunction in older adults is expected to prevent further deterioration of cerebral perfusion, recently identified as a biomarker for the very early (preclinical) stages of AD, mTOR attenuation may potentially block the initiation and progression of AD.


Subject(s)
Aging/genetics , Cerebrovascular Circulation/physiology , Cognitive Dysfunction/physiopathology , TOR Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Rats
11.
J Neurochem ; 106(1): 158-69, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18346202

ABSTRACT

It is well known that immunoproteasome generates peptides for MHC Class I occupancy and recognition by cytotoxic T lymphocytes (CTL). The present study focused on evidence for alternative roles for immunoproteasome. Retina and brain were analyzed for expression of immunoproteasome subunits using immunohistochemistry and western blotting under normal conditions and after injury/stress induced by CTL attack on glia (brain) or neurons (retina). Normal retina expressed substantial levels of immunoproteasome in glia, neurons, and retinal pigment epithelium. The basal level of immunoproteasome in retina was two-fold higher than in brain; CTL-induced retinal injury further up-regulated immunoproteasome expression. Immunoproteasome up-regulation was also observed in injured brain and corresponded with expression in Purkinje cells, microglia, astrocytes, and oligodendrocytes. These results suggest that the normal environment of the retina is sufficiently challenging to require on-going expression of immunoproteasome. Further, immunoproteasome up-regulation with retinal and brain injury implies a role in neuronal protection and/or repair of damage.


Subject(s)
Brain/immunology , Encephalitis/immunology , Proteasome Endopeptidase Complex/immunology , Retina/immunology , Retinitis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain/physiopathology , Cell Communication/immunology , Encephalitis/physiopathology , Mice , Mice, Transgenic , Nerve Degeneration/immunology , Nerve Degeneration/physiopathology , Nerve Regeneration/immunology , Neuroglia/immunology , Neuroglia/pathology , Neuronal Plasticity/immunology , Neurons/immunology , Neurons/pathology , Pigment Epithelium of Eye/immunology , Pigment Epithelium of Eye/physiopathology , Protein Subunits/immunology , Recovery of Function/immunology , Retina/physiopathology , Retinitis/physiopathology , Up-Regulation/immunology
12.
J Cereb Blood Flow Metab ; 38(1): 58-74, 2018 01.
Article in English | MEDLINE | ID: mdl-28511572

ABSTRACT

We recently showed that mTOR attenuation blocks progression and abrogates established cognitive deficits in Alzheimer's disease (AD) mouse models. These outcomes were associated with the restoration of cerebral blood flow (CBF) and brain vascular density (BVD) resulting from relief of mTOR inhibition of NO release. Recent reports suggested a role of mTOR in atherosclerosis. Because mTOR drives aging and vascular dysfunction is a universal feature of aging, we hypothesized that mTOR may contribute to brain vascular and cognitive dysfunction associated with atherosclerosis. We measured CBF, BVD, cognitive function, markers of inflammation, and parameters of cardiovascular disease in LDLR-/- mice fed maintenance or high-fat diet ± rapamycin. Cardiovascular pathologies were proportional to severity of brain vascular dysfunction. Aortic atheromas were reduced, CBF and BVD were restored, and cognitive dysfunction was attenuated potentially through reduction in systemic and brain inflammation following chronic mTOR attenuation. Our studies suggest that mTOR regulates vascular integrity and function and that mTOR attenuation may restore neurovascular function and cardiovascular health. Together with our previous studies in AD models, our data suggest mTOR-driven vascular damage may be a mechanism shared by age-associated neurological diseases. Therefore, mTOR attenuation may have promise for treatment of cognitive impairment in atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Cerebrovascular Circulation/physiology , Cognitive Dysfunction/metabolism , Dementia, Vascular/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Atherosclerosis/complications , Cognitive Dysfunction/etiology , Dementia, Vascular/etiology , Disease Models, Animal , Mice , Mice, Knockout , Receptors, LDL/deficiency
13.
FEBS Lett ; 581(5): 885-90, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17289037

ABSTRACT

The proteasome mediates pathways associated with oxidative stress and inflammation, two pathogenic events correlated with age-related macular degeneration (AMD). In human donor eyes corresponding to four stages of AMD, we found the proteasomal chymotrypsin-like activity increased in neurosensory retina with disease progression. Increased activity correlated with a dramatic increase in the inducible subunits of the immunoproteasome, which was not due to an increase in CD45 positive immune cells in the retina. The novel observation of proteasome transformation may reflect retinal response to local inflammation or oxidative stress with AMD.


Subject(s)
Macular Degeneration/enzymology , Proteasome Endopeptidase Complex/metabolism , Animals , Cytokines/metabolism , Humans , In Vitro Techniques , Inflammation/enzymology , Leukocyte Common Antigens/metabolism , Macular Degeneration/immunology , Macular Degeneration/pathology , Oxidative Stress , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/immunology , Protein Subunits , Rats , Rats, Inbred F344 , Retina/enzymology
15.
PLoS One ; 10(5): e0126768, 2015.
Article in English | MEDLINE | ID: mdl-25978061

ABSTRACT

The immunoproteasome is upregulated by disease, oxidative stress, and inflammatory cytokines, suggesting an expanded role for the immunoproteasome in stress signaling that goes beyond its canonical role in generating peptides for antigen presentation. The signaling pathways that are regulated by the immunoproteasome remain elusive. However, previous studies suggest a role for the immunoproteasome in the regulation of PTEN and NF-κB signaling. One well-known pathway upstream of NF-κB and downstream of PTEN is the Akt signaling pathway, which is responsible for mediating cellular survival and is modulated after optic nerve crush (ONC). This study investigated the role of retinal immunoproteasome after injury induced by ONC, focusing on the Akt cell survival pathway. Retinas or retinal pigment epithelial (RPE) cells from wild type (WT) and knockout (KO) mice lacking either one (LMP2) or two (LMP7 and MECL-1) catalytic subunits of the immunoproteasome were utilized in this study. We show that mRNA and protein levels of the immunoproteasome subunits are significantly upregulated in WT retinas following ONC. Mice lacking the immunoproteasome subunits show either a delayed or dampened apoptotic response as well as altered Akt signaling, compared to WT mice after ONC. Treatment of the RPE cells with insulin growth factor-1 (IGF-1) to stimulate Akt signaling confirmed that the immunoproteasome modulates this pathway, and most likely modulates parallel pathways as well. This study links the inducible expression of the immunoproteasome following retinal injury to Akt signaling, which is important in many disease pathways.


Subject(s)
Optic Nerve/metabolism , Proteasome Endopeptidase Complex/genetics , Retina/metabolism , Animals , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Epithelial Cells/metabolism , Female , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Crush/methods , Oxidative Stress/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Up-Regulation/genetics
16.
J Cereb Blood Flow Metab ; 33(9): 1412-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23801246

ABSTRACT

Vascular pathology is a major feature of Alzheimer's disease (AD) and other dementias. We recently showed that chronic administration of the target-of-rapamycin (TOR) inhibitor rapamycin, which extends lifespan and delays aging, halts the progression of AD-like disease in transgenic human (h)APP mice modeling AD when administered before disease onset. Here we demonstrate that chronic reduction of TOR activity by rapamycin treatment started after disease onset restored cerebral blood flow (CBF) and brain vascular density, reduced cerebral amyloid angiopathy and microhemorrhages, decreased amyloid burden, and improved cognitive function in symptomatic hAPP (AD) mice. Like acetylcholine (ACh), a potent vasodilator, acute rapamycin treatment induced the phosphorylation of endothelial nitric oxide (NO) synthase (eNOS) and NO release in brain endothelium. Administration of the NOS inhibitor L-NG-Nitroarginine methyl ester reversed vasodilation as well as the protective effects of rapamycin on CBF and vasculature integrity, indicating that rapamycin preserves vascular density and CBF in AD mouse brains through NOS activation. Taken together, our data suggest that chronic reduction of TOR activity by rapamycin blocked the progression of AD-like cognitive and histopathological deficits by preserving brain vascular integrity and function. Drugs that inhibit the TOR pathway may have promise as a therapy for AD and possibly for vascular dementias.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Bacterial Agents/pharmacology , Brain/metabolism , Memory/drug effects , Nitric Oxide Synthase Type III/metabolism , Sirolimus/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Transgenic , Nitric Oxide/biosynthesis , Nitric Oxide/genetics , Nitric Oxide Synthase Type III/genetics , Nitroarginine/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Vasodilation/drug effects , Vasodilation/genetics
17.
J Cereb Blood Flow Metab ; 33(10): 1605-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23838831

ABSTRACT

Recent studies have challenged the prevailing view that reduced mitochondrial function and increased oxidative stress are correlated with reduced longevity. Mice carrying a homozygous knockout (KO) of the Surf1 gene showed a significant decrease in mitochondrial electron transport chain Complex IV activity, yet displayed increased lifespan and reduced brain damage after excitotoxic insults. In the present study, we examined brain metabolism, brain hemodynamics, and memory of Surf1 KO mice using in vitro measures of mitochondrial function, in vivo neuroimaging, and behavioral testing. We show that decreased respiration and increased generation of hydrogen peroxide in isolated Surf1 KO brain mitochondria are associated with increased brain glucose metabolism, cerebral blood flow, and lactate levels, and with enhanced memory in Surf1 KO mice. These metabolic and functional changes in Surf1 KO brains were accompanied by higher levels of hypoxia-inducible factor 1 alpha, and by increases in the activated form of cyclic AMP response element-binding factor, which is integral to memory formation. These findings suggest that Surf1 deficiency-induced metabolic alterations may have positive effects on brain function. Exploring the relationship between mitochondrial activity, oxidative stress, and brain function will enhance our understanding of cognitive aging and of age-related neurologic disorders.


Subject(s)
Brain/metabolism , Cerebrovascular Circulation , Membrane Proteins/genetics , Memory/physiology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Adenosine Triphosphate/metabolism , Animals , Behavior, Animal/physiology , Blood Flow Velocity/genetics , Blood Flow Velocity/physiology , Brain/blood supply , Cerebrovascular Circulation/genetics , Glucose/metabolism , Hydrogen Peroxide/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Maze Learning/physiology , Membrane Proteins/deficiency , Mice , Mice, Knockout , Mitochondria/enzymology , Mitochondrial Proteins/deficiency , Oxygen Consumption/physiology
18.
Invest Ophthalmol Vis Sci ; 52(2): 714-23, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20881299

ABSTRACT

PURPOSE: The immunoproteasome is a proteasome subtype with a well-characterized role in the immune system. The presence of high immunoproteasome concentrations in the photoreceptors and synaptic regions of the immune-privileged retina implies a role in visual transmission. In this study, immunoproteasome knockout (KO) mice lacking either one (lmp7(-/-), L7) or two (lmp7(-/-)/mecl-1(-/-), L7M1) catalytic subunits of the immunoproteasome were used to test the hypothesis that it is essential for the maintenance of normal retinal function. METHODS: Wild-type (WT) and immunoproteasome KO mice lacking either one (L7) or two (L7M1) catalytic subunits of the immunoproteasome were studied to determine the importance of the immunoproteasome in maintaining normal retinal function and morphology. Changes in retinal morphology were assessed in mice 2 to 24 months of age. Retinal function was measured with electroretinography (ERG), and relative content of select retinal proteins was assessed by immunoblot analysis. RESULTS: Retinal morphometry showed no major abnormalities in age-matched WT or KO mice. No significant difference was observed in the levels of proteins involved in vision transmission. ERGs from KO mice exhibited an approximate 25% decrease in amplitude of the dark- and light-adapted b-waves and faster dark-adapted b-wave implicit times. CONCLUSIONS: Immunoproteasome deficiency causes defects in bipolar cell response. These results support a previously unrecognized role for the immunoproteasome in vision transmission.


Subject(s)
Cysteine Endopeptidases/physiology , Proteasome Endopeptidase Complex/physiology , Retina/physiology , Aging/physiology , Animals , Apoptosis , Catalytic Domain , Electroretinography , Fluorescent Antibody Technique, Indirect , Gene Silencing , Immune System/physiology , Immunoblotting , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/cytology , Retinal Bipolar Cells/physiology , Vision, Ocular/physiology
SELECTION OF CITATIONS
SEARCH DETAIL