Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167086, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38378084

ABSTRACT

Alzheimer's disease (AD) is characterized by the formation ß-amyloid (Aß) deposited neuritic plaques. Recent evidence suggests that abnormal lipid metabolism and accumulation could serve as biomarkers for neurodegenerative diseases, including AD. Tubular endoplasmic reticulum protein, reticulon 3 (RTN3), plays a crucial role in the development of neuritic plaque and lipid metabolism in AD brains. In present study, we sought to investigate a potential association between neutral lipid accumulation and AD pathology. BODIPY 500/510 dye was used to label neutral lipid surrounding Aß plaques in APPNL-G-F mouse and AD postmortem brains samples. Immunofluorescent images were captured using confocal microscope and co-localization between lipid metabolism proteins and neutral lipids were evaluated. Lipid accumulation in Aß plaque surrounding dystrophic neurites (DNs) was observed in the cortical region of AD mouse models and human AD brain samples. The neutral lipid staining was not co-localized with IBA1-labeled microglia or GFAP-labeled astrocytes, but it was co-labeled with VAMP2 and neurofilament. We further showed that neutral lipids were accumulated in RTN3 immunoreactive DNs. Both the neutral lipids accumulation and RIDNs formation showed age-dependent patterns in surrounding amyloid plaques. Mechanistic studies revealed that RTN3 likely contributes to the enrichment of neutral lipids near plaques by interacting with heat shock cognate protein 70 (HSC70) and diminishing its function in chaperone-mediated lipophagy. Our study provides immunohistochemical evidence of neutral lipids being enriched in DNs near amyloid plaques. Our findings shed light on RTN3-mediaed lipid accumulation in AD neuropathology and provide fresh insights into the role of RTN3 in neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Mice , Animals , Humans , Alzheimer Disease/metabolism , Neurites/pathology , Plaque, Amyloid/metabolism , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Lipids
2.
J Alzheimers Dis ; 97(1): 395-408, 2024.
Article in English | MEDLINE | ID: mdl-38160353

ABSTRACT

BACKGROUND: While symptoms related to lower urinary tract dysfunction (LUTD) are common in individuals with Alzheimer's disease (AD), pathophysiological links between AD and LUTD remain unclear. OBJECTIVE: This study aimed to investigate whether AD neuropathology would cause autonomic dysfunction along the spinal cord-bladder axis, which could result in alterations in bladder muscle kinetics. METHODS: We utilized APPNL-G-F/NL-G-F knock-in (APP KI) and APPwt/wt (wild-type) mice at two different ages, 4- and 10-month-old, to investigate how AD impacts bladder tissue function by immunohistochemistry, western blotting, and pharmacomyography. RESULTS: We showed that the mucosal layer partially separated from the detrusor in 10-month-old APP KI mouse bladders. Although there was no detectable amyloid deposition in the APP KI bladder, we found amyloid plaques in APP KI lumbar spinal cord. Further immunoblot analysis revealed that tyrosine hydroxylase protein levels were significantly reduced in both 4- and 10-month-old bladder tissues, suggesting reduction of norepinephrine synthesis in APP KI mouse bladders. In contrast, the level of ß2 adrenergic receptor was increased in 4-month-old but not 10-month-old APP KI bladders. In bladder strips, the adrenergic agonist isoproterenol induced increased relaxation in 4- but not 10-month-old APP KI bladders. With 10 Hz electrical field stimulation, 10-month-old APP KI bladder strips were more responsive than wild-type controls, with no differences observed in 4-month-old APP KI bladders. CONCLUSIONS: APP KI mice exhibit LUTD, which is likely arising from amyloid pathology in the spinal cord, and results in maturational declines in presynaptic activity combined with compensatory postsynaptic upregulation.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Disease Models, Animal , Mice, Transgenic , Plaque, Amyloid/pathology
3.
J Neurosci ; 43(35): 6197-6211, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37536983

ABSTRACT

Alzheimer's disease (AD) increases the risk for seizures and sleep disorders. We show here that germline deletion of ß-site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) in neurons, but not in astrocytes, increased epileptiform activity. However, Bace1 deletion at adult ages did not alter the normal EEG waveform, indicating less concern for BACE1 inhibition in patients. Moreover, we showed that deletion of Bace1 in the adult was able to reverse epileptiform activity in 5xFAD mice. Intriguingly, treating 5xFAD and APPNL-G-F/NL-G-F (APP KI) mice of either sex with one BACE1 inhibitor Lanabecestat (AZD3293) dramatically increased epileptiform spiking, likely resulting from an off-target effect. We also monitored sleep-wake pathologies in these mice and showed increased wakefulness, decreased non-rapid eye movement sleep, and rapid eye movement sleep in both 5xFAD and APP KI mice; BACE1 inhibition in the adult 5xFAD mice reversed plaque load and sleep disturbances, but this was not seen in APP KI mice. Further studies with and without BACE1 inhibitor treatment showed different levels of plaque-associated microgliosis and activated microglial proteins in 5xFAD mice compared with APP KI mice. Together, BACE1 inhibition should be developed to avoid off-target effect for achieving benefits in reducing epileptic activity and sleep disturbance in Alzheimer's patients.SIGNIFICANCE STATEMENT BACE1 is widely recognized as a therapeutic target for treating Alzheimer's disease patients. However, BACE1 inhibitors failed in clinical trials because of inability to show cognitive improvement in patients. Here we show that BACE1 inhibition actually reduces sleep disturbances and epileptic seizures; both are seen in AD patients. We further showed that one of clinically tested BACE1 inhibitors does have off-target effects, and development of safer BACE1 inhibitors will be beneficial to AD patients. Results from this study will provide useful guidance for additional drug development.


Subject(s)
Alzheimer Disease , Sleep Wake Disorders , Mice , Animals , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Mice, Transgenic , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Plaque, Amyloid , Seizures , Sleep Wake Disorders/etiology , Sleep Wake Disorders/genetics , Sleep , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Peptides/metabolism , Disease Models, Animal
4.
Hum Mol Genet ; 32(16): 2587-2599, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37228035

ABSTRACT

Reticulon (RTN) proteins are a family of proteins biochemically identified for shaping tubular endoplasmic reticulum, a subcellular structure important for vesicular transport and cell-to-cell communication. In our recent study of mice with knockout of both reticulon 1 (Rtn1) and Rtn3, we discovered that Rtn1-/-;Rtn3-/- (brief as R1R3dKO) mice exhibited neonatal lethality, despite the fact that mice deficient in either RTN1 or RTN3 alone exhibit no discernible phenotypes. This has been the first case to find early lethality in animals with deletion of partial members of RTN proteins. The complete penetrance for neonatal lethality can be attributed to multiple defects including the impaired neuromuscular junction found in the diaphragm. We also observed significantly impaired axonal growth in a regional-specific manner, detected by immunohistochemical staining with antibodies to neurofilament light chain and neurofilament medium chain. Ultrastructural examination by electron microscopy revealed a significant reduction in synaptic active zone length in the hippocampus. Mechanistic exploration by unbiased proteomic assays revealed reduction of proteins such as FMR1, Staufen2, Cyfip1, Cullin-4B and PDE2a, which are known components in the fragile X mental retardation pathway. Together, our results reveal that RTN1 and RTN3 are required to orchestrate neurofilament organization and intact synaptic structure of the central nervous system.


Subject(s)
Axons , Cytoskeleton , Hippocampus , Nerve Tissue Proteins , Animals , Mice , Genes, Lethal , Mice, Knockout , Axons/metabolism , Axons/pathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Nerve Tissue Proteins/metabolism , Endoplasmic Reticulum/metabolism , Synapses , Hippocampus/metabolism , Hippocampus/pathology
5.
Hum Mol Genet ; 32(7): 1193-1207, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36370042

ABSTRACT

Beta amyloid cleaving enzyme 1 (BACE1) is largely expressed by neurons and is the sole ß-secretase for initiating the production of neuronal ß-amyloid peptides (Aß). To fully understand the physiological functions of neuronal BACE1, we used mouse genetic approach coupled with unbiased single nucleus RNA sequencing (snRNAseq) to investigate how targeted deletion of Bace1 in neurons, driven by Thy-1-Cre recombinase, would affect functions in the nervous system. Our transcriptome results revealed that BACE1 is essential for maturation of neural precursor cells and oligodendrocytes in mice. RNA velocity analysis confirmed deficit in the trajectory of neuroblasts in reaching the immature granule neuron state in young Bace1fl/fl; Thy1-cre mice. Further analysis of differential gene expression indicated changes in genes important for SNARE signaling, tight junction signaling, synaptogenesis and insulin secretion pathways. Morphological studies revealed a hypomyelination in Bace1fl/fl;Thy1-cre sciatic nerves, but no detectable myelination changes in the corpus callosum, despite clear reduction in myelination proteins in the brain. Functional studies showed reduction in long-term potential, defects in synaptogenesis and learning behavioral. Altogether, our results show that neuronal BACE1 is critical for optimal development of central and peripheral nervous system, and inhibition of neuronal BACE1 will result in deficits in synaptic functions and cognitive behaviors.


Subject(s)
Alzheimer Disease , Neural Stem Cells , Mice , Animals , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Amyloid beta-Peptides/metabolism , Oligodendroglia/metabolism , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/metabolism
6.
Tree Physiol ; 43(3): 452-466, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36263985

ABSTRACT

A cultivable endophytic fungus, Piriformospora indica, improves growth and enhances stress tolerance of host plants, but the underlying mechanisms remain unknown. We hypothesized that P. indica enhanced the drought tolerance of the host by regulating the antioxidant defense system and composition of fatty acids. Trifoliate orange (Poncirus trifoliata) seedlings were inoculated with P. indica under ample water and drought stress to analyze the change in plant growth, reactive oxygen species (ROS) levels, antioxidant enzyme activities, non-enzymatic antioxidant concentrations, fatty acid compositions, and expressions of both antioxidant enzyme genes and fatty acid desaturase (FAD) genes. The 9-week soil water deficit significantly increased the colonization of P. indica to roots, and P. indica promoted the increase of shoot biomass under drought. Soil drought triggered an elevation of hydrogen peroxide in roots, while the inoculated plants had lower levels of ROS (hydrogen peroxide and superoxide anion radicals) and lower degree of membrane lipid peroxidation (based on malondialdehyde levels) under drought. Drought treatment also elevated ascorbic acid and glutathione concentrations, and the elevation was further amplified after P. indica inoculation. Inoculated plants under drought also recorded significantly higher iron-superoxide dismutase (Fe-SOD), manganese-superoxide dismutase (Mn-SOD), peroxidases, catalase, glutathione reductase and ascorbate peroxidase activities, accompanied by up-regulation of PtFe-SOD and PtCu/Zn-SOD expressions. Inoculation with P. indica significantly increased total saturated fatty acids (e.g., C6:0, C15:0, C16:0, C23:0 and C24:0) concentration and reduced total unsaturated fatty acids (e.g., C18:1N9C, C18:2N6, C18:3N3, C18:1N12 and C19:1N9T) concentrations, leading to a decrease in the unsaturation index of fatty acids, which may be associated with the up-regulation of PtFAD2 and PtFAD6 and down-regulation of PtΔ9. It was concluded that the colonization of P. indica can activate enzyme and non-enzyme defense systems and regulate the composition of fatty acids under drought, thus alleviating the oxidative damage to the host caused by drought.


Subject(s)
Basidiomycota , Poncirus , Antioxidants/metabolism , Poncirus/metabolism , Reactive Oxygen Species/metabolism , Drought Resistance , Hydrogen Peroxide/metabolism , Fatty Acids/metabolism , Basidiomycota/physiology , Superoxide Dismutase/metabolism , Droughts , Water/metabolism
7.
Exp Mol Med ; 54(5): 653-661, 2022 05.
Article in English | MEDLINE | ID: mdl-35596061

ABSTRACT

Reticulon 3 (RTN3) is an endoplasmic reticulum protein that has previously been shown to play roles in neurodegenerative diseases, but little is known about its function in the kidneys. The aim of the present study was to clarify the roles of RTN3 in chronic kidney disease (CKD) and kidney fibrosis. In this study, RTN3 levels were measured in kidney tissues from healthy controls and CKD or kidney fibrosis patients. An RTN3-null mouse model was generated to explore the pathophysiological roles of RTN3 in the kidneys. The underlying mechanisms were studied in primary proximal tubular epithelial cells and HEK293 cells in vitro. The results showed that (1) a reduction in RTN3 in mice induces CKD and kidney fibrosis; (2) decreased RTN3 expression is found in patients with CKD; (3) RTN3 plays critical roles in regulating collagen biosynthesis and mitochondrial function; and (4) mechanistically, RTN3 regulates these phenotypes by interacting with GC-Rich Promoter Binding Protein 1 (GPBP1), which activates the IGF2-JAK2-STAT3 pathway. Our study indicates that RTN3 might play crucial roles in CKD and kidney fibrosis and that a reduction in RTN3 in the kidneys might be a risk factor for CKD and kidney fibrosis.


Subject(s)
Membrane Proteins , Nerve Tissue Proteins , Renal Insufficiency, Chronic , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins , Epithelial Cells/metabolism , Fibrosis , HEK293 Cells , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phenotype , Renal Insufficiency, Chronic/genetics
8.
Neurobiol Aging ; 107: 21-29, 2021 11.
Article in English | MEDLINE | ID: mdl-34371284

ABSTRACT

Formation of Reticulon 3 (RTN3)-immunoreactive dystrophic neurites (RIDNs) occurs early during the growth of amyloid plaques in Alzheimer's disease (AD) brains. We have shown that RIDNs in AD and aging mouse brains are composed of abnormally clustered tubular endoplasmic reticulum (ER) and degenerating mitochondria. To understand RTN3-mediated abnormal tubular ER clustering, we aimed to identify proteins that interact with RTN3 and impact accumulation of tubular ER in RIDNs. We found that the N-terminal domain of RTN3, which is unique among RTN family members, specifically interacted with dynactin 6 (DCTN6), a protein involved in dynein-mediated retrograde transport of cargo vesicles. DCTN6 protein levels decrease with aging in the hippocampal regions of WT mice. We found that DCTN6 deficiency enhanced RTN3 protein levels, high molecular weight RTN3 levels, and hippocampus-specific RIDN formation in aging brains of transgenic mice overexpressing RTN3. Our results suggest that the DCTN6-RTN3 interaction mediates tubular ER trafficking in axons, and a DCTN6 deficiency in the hippocampus impairs axonal ER trafficking, leading to abnormal ER accumulation and RIDN formation in brains of aging mice.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Dynactin Complex/deficiency , Neurites/pathology , Neuroaxonal Dystrophies/etiology , Aging/metabolism , Alzheimer Disease/metabolism , Animals , Axonal Transport , Dynactin Complex/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/metabolism
9.
Mol Psychiatry ; 26(11): 6394-6410, 2021 11.
Article in English | MEDLINE | ID: mdl-34158621

ABSTRACT

BACE1 initiates production of ß-amyloid peptides (Aß), which is associated with cognitive dysfunction in Alzheimer's disease (AD) due to abnormal oligomerization and aggregation. While BACE1 inhibitors show strong reduction in Aß deposition, they fail to improve cognitive function in patients, largely due to its role in synaptic function. We show that BACE1 is required for optimal release of synaptic vesicles. BACE1 deficiency or inhibition decreases synaptic vesicle docking in the synaptic active zones. Consistently, BACE1-null mice or mice treated with clinically tested BACE1 inhibitors Verubecestat and Lanabecestat exhibit severe reduction in hippocampal LTP and learning behaviors. To counterbalance this synaptic deficit, we discovered that BACE1-null mice treated with positive allosteric modulators (PAMs) of metabotropic glutamate receptor 1 (mGluR1), whose levels were reduced in BACE1-null mice and significantly improved long-term potentiation and cognitive behaviors. Similarly, mice treated with mGluR1 PAM showed significantly mitigated synaptic deficits caused by BACE1 inhibitors. Together, our data suggest that a therapy combining BACE1 inhibitors for reducing amyloid deposition and an mGluR1 PAM for counteracting BACE1-mediated synaptic deficits appears to be an effective approach for treating AD patients.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Animals , Aspartic Acid Endopeptidases , Humans , Mice , Synaptic Vesicles
10.
J Neurosci ; 40(5): 1133-1144, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31822518

ABSTRACT

Neurofibrillary tangles likely cause neurodegeneration in Alzheimer's disease (AD). We demonstrate that the CX3CL1 C-terminal domain can upregulate neurogenesis, which may ameliorate neurodegeneration. Here we generated transgenic (Tg-CX3CL1) mice by overexpressing CX3CL1 in neurons. Tg-CX3CL1 mice exhibit enhanced neurogenesis in both subgranular and subventricular zones. This enhanced neurogenesis correlates well with elevated expression of TGF-ß2 and TGF-ß3, and activation of their downstream signaling molecule Smad2. Intriguingly, the enhanced adult neurogenesis was mitigated when Smad2 expression was deleted in neurons, supporting a role for the CX3CL1-TGF-ß2/3-Smad2 pathway in the control of adult neurogenesis. When Tg-CX3CL1 mice were crossed with Alzheimer's PS19 mice, which overexpress a tau P301S mutation and exhibit age-dependent neurofibrillary tangles and neurodegeneration, overexpressed CX3CL1 in both male and female mice was sufficient to rescue the neurodegeneration, increase survival time, and improve cognitive function. Hence, we provide in vivo evidence that CX3CL1 is a strong activator of adult neurogenesis, and that it reduces neuronal loss and improves cognitive function in AD.SIGNIFICANCE STATEMENT This study will be the first to demonstrate that enhanced neurogenesis by overexpressed CX3CL1 is mitigated by disruption of Smad2 signaling and is independent of its interaction with CX3CR1. Overexpression of CX3CL1 lengthens the life span of PS19 tau mice by enhancing adult neurogenesis while having minimal effect on tau pathology. Enhancing neuronal CX3CL1, mainly the C-terminal fragment, is a therapeutic strategy for blocking or reversing neuronal loss in Alzheimer's disease or related neurodegenerative disease patients.


Subject(s)
Alzheimer Disease , Chemokine CX3CL1/metabolism , Neurogenesis , Neurons/metabolism , Smad2 Protein/metabolism , Spatial Memory/physiology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Disease Models, Animal , Female , Male , Mice, Transgenic , Neurons/pathology
11.
Open Med (Wars) ; 14: 625-628, 2019.
Article in English | MEDLINE | ID: mdl-31535033

ABSTRACT

BACKGROUND: Interleukin-33 (IL-33) and homocysteine (Hcy) were found to be up-regulated in patients with diabetic nephropathy (DN), and the present study aimed to investigate whether metformin (MT) can influence the serum levels of IL-33 and Hcy in patients with DN. METHODS: Sixty patients with type 2 diabetes mellitus (DM) were divided into DM group (albumin: Alb <20 mg/L), DN group (Alb >20mg/L), and DN+ MT treatment group, with 20 cases in each group. Patients in each group were treated with insulin for 3 months, and patients in DN+MT group was treated with insulin+MT for 3 months. The serum levels of IL-33, urinary microalbumin excretion rate (UAE), body mass index (BMI), total cholesterol (TC), high density lipoprotein cholesterol (HDL-C), creatinine (Cr), cystatin C (CysC) and Hcy were measured before and after medication. Twenty normal subjects were involved as control. RESULTS: BMI, Hcy and TC were reduced and HDL-C was increased of patients had been treated with metformin and insulin. UAE, Cr, Ccr and CysC had no differences before and after treatment. The serum level of IL-33 significantly up-regulated in patients with DN, and MT treatment significantly decreased the serum level of IL-33 in patients with DN. CONCLUSION: Metformin could alleviate the symptom of patient with DN through decreasing the serum level of IL-33 and Hcy.

12.
J Exp Med ; 216(8): 1891-1903, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31209068

ABSTRACT

The membrane-anchored CX3CL1 is best known to exert its signaling function through binding its receptor CX3CR1. This study demonstrates a novel function that CX3CL1 exerts. CX3CL1 is sequentially cleaved by α-, ß-, and γ-secretase, and the released CX3CL1 intracellular domain (CX3CL1-ICD) would translocate into the cell nucleus to alter gene expression due to this back-signaling function. Amyloid deposition and neuronal loss were significantly reduced when membrane-anchored CX3CL1 C-terminal fragment (CX3CL1-ct) was overexpressed in Alzheimer's 5xFAD mouse model. The reversal of neuronal loss in 5xFAD can be attributed to increased neurogenesis by CX3CL1-ICD, as revealed by morphological and unbiased RNA-sequencing analyses. Mechanistically, this CX3CL1 back-signal likely enhances developmental and adult neurogenesis through the TGFß2/3-Smad2/3 pathway and other genes important for neurogenesis. Induction of CX3CL1 back-signaling may not only be a promising novel mechanism to replenish neuronal loss but also for reducing amyloid deposition for Alzheimer's treatment.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Chemokine CX3CL1/metabolism , Neurogenesis/genetics , Plaque, Amyloid/metabolism , Protein Domains/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cell Nucleus/metabolism , Chemokine CX3CL1/chemistry , Chemokine CX3CL1/genetics , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport , Transcriptional Activation/genetics , Transfection
13.
Circulation ; 138(17): 1828-1838, 2018 10 23.
Article in English | MEDLINE | ID: mdl-29716941

ABSTRACT

BACKGROUND: Reticulon 3 (RTN3) is an endoplasmic reticulum protein that has previously been shown to play a role in neurodegenerative diseases, but little is known about its role in lipid metabolism. METHODS: Obese patients (n=149), hypertriglyceridemic patients (n=343), and healthy control subjects (n=84) were enrolled to assess their levels of RTN3. To explore the pathophysiological roles of RTN3 in the control of lipid metabolism, we used transgenic mice overexpressing the wild-type human RTN3 gene, the RTN3-null transgenic mouse model, and multiple Caenorhabditis legans strains for molecular characterization. The underlying mechanisms were studied with 3T3L1 cell cultures in vitro. RESULTS: We report that overexpressed RTN3 in mice induces obesity and higher accumulation of triglycerides. Increased RTN3 expression is also found in patients with obesity and hypertriglyceridemia. We reveal that RTN3 plays critical roles in regulating the biosynthesis and storage of triglycerides and in controlling lipid droplet expansion. Mechanistically, RTN3 regulates these events through its interactions with heat shock protein family A (Hsp70) member 5, and this enhanced interaction increases sterol regulatory element-binding protein 1c and AMP-activated kinase activity. CONCLUSIONS: This study provides evidence for a role of RTN3 in inducing obesity and triglyceride accumulation and suggests that inhibiting the expression of RTN3 in fat tissue may be a novel therapeutic approach to treat obesity and hypertriglyceridemia.


Subject(s)
Adipose Tissue/metabolism , Carrier Proteins/blood , Heat-Shock Proteins/metabolism , Hypertriglyceridemia/blood , Membrane Proteins/blood , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/metabolism , Obesity/blood , Triglycerides/blood , 3T3-L1 Cells , AMP-Activated Protein Kinases , Adolescent , Adult , Animals , Biomarkers/blood , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Carrier Proteins/genetics , Case-Control Studies , Endoplasmic Reticulum Chaperone BiP , Female , Genetic Predisposition to Disease , Heat-Shock Proteins/genetics , Humans , Hypertriglyceridemia/diagnosis , Hypertriglyceridemia/genetics , Lipid Droplets/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Middle Aged , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Obesity/diagnosis , Obesity/genetics , Phenotype , Sterol Regulatory Element Binding Protein 1/metabolism , Up-Regulation , Young Adult
14.
J Exp Med ; 215(3): 927-940, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29444819

ABSTRACT

BACE1 initiates the generation of the ß-amyloid peptide, which likely causes Alzheimer's disease (AD) when accumulated abnormally. BACE1 inhibitory drugs are currently being developed to treat AD patients. To mimic BACE1 inhibition in adults, we generated BACE1 conditional knockout (BACE1fl/fl) mice and bred BACE1fl/fl mice with ubiquitin-CreER mice to induce deletion of BACE1 after passing early developmental stages. Strikingly, sequential and increased deletion of BACE1 in an adult AD mouse model (5xFAD) was capable of completely reversing amyloid deposition. This reversal in amyloid deposition also resulted in significant improvement in gliosis and neuritic dystrophy. Moreover, synaptic functions, as determined by long-term potentiation and contextual fear conditioning experiments, were significantly improved, correlating with the reversal of amyloid plaques. Our results demonstrate that sustained and increasing BACE1 inhibition in adults can reverse amyloid deposition in an AD mouse model, and this observation will help to provide guidance for the proper use of BACE1 inhibitors in human patients.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Amyloid/metabolism , Aspartic Acid Endopeptidases/deficiency , Cognition , Gene Deletion , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Behavior, Animal , Disease Models, Animal , Integrases/metabolism , Learning , Long-Term Potentiation , Mice, Transgenic , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology
15.
Hum Genet ; 137(2): 141-150, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29356939

ABSTRACT

Reticulon 3 (RTN3) is a neuronally-expressed reticulon family protein that was previously shown to negatively regulate BACE1, a protease that is required for the generation of ß-amyloid peptides (Aß) from amyloid precursor protein. Despite biochemical and morphological evidence that supports a role of RTN3 in the formation of neuritic amyloid plaques, no systematic analyses of RTN3 mutations in patients with Alzheimer's disease (AD) have yet been reported. RTN3 were targeted sequenced in 154 sporadic early-onset and 285 late-onset AD patients. Luciferase reporter assay and kymographs were performed to analysis the expression of RNT3 and BACE1-RFP particle mobility on cells transfected with wild-type or variants RTN3 constructs. We identified heterozygous variants such as c.-8G > T, c.17C > A, c.42C > T, and c.116C > T from patients in the early-onset AD group and c.-8G > T, c.17C > A, from patients in the late-onset AD group. Such variants of RTN3 were not observed in control individuals. Further biochemical studies show that the RTN3 c.-8G > T variant in the 5'-untranslated region appears to cause reduced expression of RTN3. The RTN3 c.116 C > T variant causes a change of codon T39 to M39 (T39 M). Overexpression of RTN3 T39 M in cultured neurons led to impaired axonal transport of BACE1. The variants found in this study are likely genetic modifiers for RTN3-mediated formation of neuritic plaques in AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Aspartic Acid Endopeptidases/genetics , Carrier Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Adult , Aged , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Axonal Transport/genetics , Brain/metabolism , Brain/pathology , Female , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Neurons/metabolism , Neurons/physiology
16.
Stem Cell Reports ; 9(1): 217-230, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28669600

ABSTRACT

BACE1 is validated as Alzheimer's ß-secretase and a therapeutic target for Alzheimer's disease. In examining BACE1-null mice, we discovered that BACE1 deficiency develops abnormal clusters of immature neurons, forming doublecortin-positive neuroblasts, in the developing dentate gyrus, mainly in the subpial zone (SPZ). Such clusters were rarely observed in wild-type SPZ and not reported in other mouse models. To understand their origins and fates, we examined how neuroblasts in BACE1-null SPZ mature and migrate during early postnatal development. We show that such neuroblasts are destined to form Prox1-positive granule cells in the dentate granule cell layer, and mainly mature to form excitatory neurons, but not inhibitory neurons. Mechanistically, higher levels of reelin potentially contribute to abnormal neurogenesis and timely migration in BACE1-null SPZ. Altogether, we demonstrate that BACE1 is a critical regulator in forming the dentate granule cell layer through timely maturation and migration of SPZ neuroblasts.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Dentate Gyrus/pathology , Gene Deletion , Neurons/pathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Dentate Gyrus/metabolism , Extracellular Matrix Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis , Neurons/metabolism , Reelin Protein , Serine Endopeptidases/metabolism
17.
Sci Rep ; 7(1): 6145, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28733667

ABSTRACT

Reticulon proteins (RTNs), consisting of RTN1 to RTN4, were previously shown to interact with BACE1 by negatively modulating its secretase activity. In RTN3-null mice, RTN1 expression was slightly elevated. To understand the in vivo role of RTN1, we generated RTN1-null mice and compared the effects of RTN1 and RTN3 on BACE1 modulation. We show that RTN1 is mostly expressed by neurons and not by glial cells under normal conditions, similar to the expression of RTN3. However, RTN1 is more localized in dendrites and is an excellent marker for dendrites of Purkinje cells, while RTN3 expression is less evident in dendrites. This differential localization also correlates with their associations with amyloid plaques in Alzheimer's brains: RTN3, but not RTN1, is abundantly enriched in dystrophic neurites. RTN3 deficiency causes elevation of BACE1 protein levels, while RTN1 deficiency shows no obvious effects on BACE1 activity due to compensation by RTN3, as RTN1 deficiency causes elevation of RTN3 expression. Hence, expression of RTN1 and RTN3 is tightly regulated in mouse brains. Together, our data show that RTN1 and RTN3 have differential effects on the formation of senile plaques in Alzheimer's brains and that RTN3 has a more prominent role in Alzheimer's pathogenesis.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Carrier Proteins/genetics , Dendrites/metabolism , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Neurons/metabolism , Plaque, Amyloid/genetics , Purkinje Cells/metabolism
18.
Glia ; 65(5): 712-726, 2017 05.
Article in English | MEDLINE | ID: mdl-28191691

ABSTRACT

BACE1 is an indispensable enzyme for generating ß-amyloid peptides, which are excessively accumulated in brains of Alzheimer's patients. However, BACE1 is also required for proper myelination of peripheral nerves, as BACE1-null mice display hypomyelination. To determine the precise effects of BACE1 on myelination, here we have uncovered a role of BACE1 in the control of Schwann cell proliferation during development. We demonstrate that BACE1 regulates the cleavage of Jagged-1 and Delta-1, two membrane-bound ligands of Notch. BACE1 deficiency induces elevated Jag-Notch signaling activity, which in turn facilitates proliferation of Schwann cells. This increase in proliferation leads to shortened internodes and decreased Schmidt-Lanterman incisures. Functionally, evoked compound action potentials in BACE1-null nerves were significantly smaller and slower, with a clear decrease in excitability. BACE1-null nerves failed to effectively use lactate as an alternative energy source under conditions of increased physiological activity. Correlatively, BACE1-null mice showed reduced performance on rotarod tests. Collectively, our data suggest that BACE1 deficiency enhances proliferation of Schwann cell due to the elevated Jag1/Delta1-Notch signaling, but fails to myelinate axons efficiently due to impaired the neuregulin1-ErbB signaling, which has been documented.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cell Proliferation/physiology , Schwann Cells/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Axons/metabolism , Cell Proliferation/genetics , Mice, Knockout , Myelin Sheath/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Schwann Cells/cytology , Sciatic Nerve/metabolism , Signal Transduction/physiology
19.
Water Sci Technol ; 74(8): 1936-1945, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27789894

ABSTRACT

Through sol-gel and dip-coating processes, commercial polyurethane sponge modified by polysiloxane was fabricated under low temperature (60 °C) and atmosphere. The contact angle of the obtained polysiloxane/polyurethane sponge is 145 ± 5°. Hence, the polysiloxane/polyurethane sponge could float on water and selectively absorb organics from the surface of the water, indicating simultaneous properties of hydrophobicity and oleophilicity. The absorbent maximum value is 50-150 times of its own weight. The polysiloxane/polyurethane sponge exhibited excellent recyclability, which could be reused by squeezing the sponge due to its high mechanical stability and flexibility. Thermogravimetry-differential thermal analysis test indicated that the polysiloxane/polyurethane sponge exhibited good thermal stability and the stable contact angle of samples tested under increasing temperature indicated its good weather resistance. Due to the commercial property of polyurethane sponge and easy-handling of polysiloxane, the polysiloxane/polyurethane sponge can be easily scaled up to recover a large-area oil spill in water and further work based on the designed equipment has been under consideration.


Subject(s)
Petroleum Pollution/prevention & control , Polyurethanes/chemistry , Recycling , Siloxanes/chemistry , Waste Disposal, Fluid/methods , Adsorption , Hydrophobic and Hydrophilic Interactions , Waste Disposal, Fluid/economics
20.
Water Sci Technol ; 73(5): 1098-103, 2016.
Article in English | MEDLINE | ID: mdl-26942532

ABSTRACT

Oil-water separation has recently become a worldwide challenge due to the frequent occurrence of oil spill accidents and increasing industrial oily wastewater. In this work, the multifunctional mesh films with underwater oleophobicity and certain bacteriostatic effects are prepared by layer-by-layer assembly of graphene oxide-silica coatings on stainless steel mesh. The mesh film exhibits excellent environmental stability under a series of harsh conditions. The new, facile and reusable separation system is proposed to achieve deep treatment of oily wastewater, and the oil collection rate can reach over 99%.


Subject(s)
Graphite/chemistry , Membranes, Artificial , Petroleum/analysis , Silicon Dioxide/chemistry , Wastewater/chemistry , Anti-Bacterial Agents , Industrial Waste , Stainless Steel , Waste Disposal, Fluid/methods , Water Pollutants, Chemical/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...