Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Poult Sci ; 103(2): 103301, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38113704

ABSTRACT

Ducks infected with duck circovirus (DuCV) show symptoms such as feather loss, growth retardation and low body weight in the flock. The virus induces immunosuppression and increases the prevalence of infection with other pathogens. However, most studies on duck circovirus were focused on coinfection, and fewer studies had been conducted on the pathogenicity of duck circovirus alone. The aim of the present study was to investigate the pathogenesis of DuCV-1 in experimentally infected specific pathogen-free ducks. In this study, we sequenced the whole genome of a strain of duck circovirus and identified the virus genotype as DuCV-1b. This strain of duck circovirus was named SDLH(OR567883). Animal pathogenicity experiments were then conducted, wherein specific pathogen-free ducks were infected by mucosal injection and abdominal injection. Infected ducks were sampled for 4 consecutive weeks after infection and showed symptoms of dwarfism. We further examined the replication of DuCV-1 in the ducks. The highest virus titers in the 2 infection groups were found in the liver and spleen, with different results for the different routes of infection. Pathological sections of duck organs were made and it was found that organs such as the liver and spleen were damaged by DuCV-1. In conclusion, our experimental results indicate that DuCV-1 can infect ducks individually and cause widespread organ damage in infected ducks.


Subject(s)
Circoviridae Infections , Circovirus , Poultry Diseases , Animals , Virulence , Chickens/genetics , Base Sequence , Circovirus/genetics , Circoviridae Infections/veterinary
2.
Sensors (Basel) ; 22(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35161463

ABSTRACT

To find an economical solution to infer the depth of the surrounding environment of unmanned agricultural vehicles (UAV), a lightweight depth estimation model called MonoDA based on a convolutional neural network is proposed. A series of sequential frames from monocular videos are used to train the model. The model is composed of two subnetworks-the depth estimation subnetwork and the pose estimation subnetwork. The former is a modified version of U-Net that reduces the number of bridges, while the latter takes EfficientNet-B0 as its backbone network to extract the features of sequential frames and predict the pose transformation relations between the frames. The self-supervised strategy is adopted during the training, which means the depth information labels of frames are not needed. Instead, the adjacent frames in the image sequence and the reprojection relation of the pose are used to train the model. Subnetworks' outputs (depth map and pose relation) are used to reconstruct the input frame, then a self-supervised loss between the reconstructed input and the original input is calculated. Finally, the loss is employed to update the parameters of the two subnetworks through the backward pass. Several experiments are conducted to evaluate the model's performance, and the results show that MonoDA has competitive accuracy over the KITTI raw dataset as well as our vineyard dataset. Besides, our method also possessed the advantage of non-sensitivity to color. On the computing platform of our UAV's environment perceptual system NVIDIA JETSON TX2, the model could run at 18.92 FPS. To sum up, our approach provides an economical solution for depth estimation by using monocular cameras, which achieves a good trade-off between accuracy and speed and can be used as a novel auxiliary depth detection paradigm for UAVs.


Subject(s)
Neural Networks, Computer , Supervised Machine Learning , Farms
3.
Front Vet Sci ; 7: 614854, 2020.
Article in English | MEDLINE | ID: mdl-33585604

ABSTRACT

Avian leukosis virus (ALV) continues evolving to obtain new genomic characters to enhance its pathogenicity. In the present study, an ALV-J strain LH20180301 was isolated from broiler breeder chickens that reached the speak of paralyzation before 20-week-old. The necropsy chickens showed subcutaneous and muscular hemorrhage, and developed tumors in multiple organs including bone, liver, spleen, and kidney. The complete provirus was then cloned and sequenced to investigate the molecular characteristics and oncogenicity etiology of this virus associated with the outbreak of disease. The genomic structure of the reported ALV-J strain LH20180301 was highly conservative with other ALVs. Recombination events between the virus with endogenous virus were identified in the viral genome. Compared with the ALV-J original HPRS-103 strain, the major recombination sites of the viral genome with ev-1 were located in 5' UTR-gag and 3' UTR regions. Phylogenetic analysis of group specific antigen gp85 encoding protein showed that the LH20180301 branched with ALV-J prevalent in "yellow chickens" of local breeds in South China. Nine amino acids (N58, D60, K70, A71, K108, N112, N113, N121, R272) in the gp85 were highly conserved among ALV-J isolates before 2012, but various mutations were found in the late isolates including LH20180301. In addition, the LH20180301 strain also had the same deletion pattern of 3' UTR with them. Therefore, LH20180301 might derive from the same ancestor with those viruses and may be the trend of ALV-J evolution in China. The defined new genomic characters in the gp85 and 3' UTR region of ALV-J might provide the molecular basis for its enhanced oncogenicity.

4.
Exp Eye Res ; 178: 228-237, 2019 01.
Article in English | MEDLINE | ID: mdl-29608906

ABSTRACT

This study evaluated the effects of elevated homocysteine (Hcy) on the oxidative stress response in retinal Müller glial cells. Elevated Hcy has been implicated in retinal diseases including glaucoma and optic neuropathy, which are characterized by retinal ganglion cell (RGC) loss. To understand the mechanisms of Hcy-induced RGC loss, in vitro and in vivo models have been utilized. In vitro isolated RGCs are quite sensitive to elevated Hcy levels, while in vivo murine models of hyperhomocysteinemia (HHcy) demonstrate a more modest RGC loss (∼20%) over a period of many months. This differential response to Hcy between isolated cells and the intact retina suggests that the retinal milieu invokes mechanisms that buffer excess Hcy. Oxidative stress has been implicated as a mechanism of Hcy-induced neuron loss and NRF2 is a transcription factor that plays a major role in regulating cytoprotective responses to oxidative stress. In the present study we investigated whether HHcy upregulates NRF2-mediated stress responses in Müller cells, the chief retinal glial cell responsible for providing trophic support to retinal neurons. Primary Müller cells were exposed to L-Hcy-thiolactone [50µM-10mM] and assessed for viability, reactive oxygen species (ROS), and glutathione (GSH) levels. Gene/protein levels of Nrf2 and levels of NRF2-regulated antioxidants (NQO1, CAT, SOD2, HMOX1, GPX1) were assessed in Hcy-exposed Müller cells. Unlike isolated RGCs, isolated Müller cells are viable over a wide range of Hcy concentrations [50 µM - 1 mM]. Moreover, when exposed to elevated Hcy, Müller cells demonstrate decreased oxidative stress and decreased ROS levels. GSH levels increased by ∼20% within 24 h exposure to Hcy. Molecular analyses revealed 2-fold increase in Nrf2 expression. Expression of antioxidant genes Nqo1, Cat, Sod2, Hmox1, Gpx1 increased significantly. The consequences of Hcy exposure were evaluated also in Müller cells harvested from Nrf2-/- mice. In contrast to WT Müller cells, in which oxidative stress decreased upon exposure to Hcy, the Nrf2-/- Müller cells showed a significant increase in oxidative stress. Our data suggest that at least during early stages of Hhcy, a cytoprotective response may be in place, mediated in part by NRF2 in Müller cells.


Subject(s)
Ependymoglial Cells/drug effects , Homocysteine/analogs & derivatives , NF-E2-Related Factor 2/metabolism , Radiation-Protective Agents/pharmacology , Animals , Antioxidant Response Elements/physiology , Cell Survival , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Glutathione/metabolism , Homocysteine/pharmacology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Up-Regulation
5.
Cell Death Dis ; 9(10): 1001, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254218

ABSTRACT

The lack of effective therapies to limit neurovascular injury in ischemic retinopathy is a major clinical problem. This study aimed to examine the role of ureohydrolase enzyme, arginase 1 (A1), in retinal ischemia-reperfusion (IR) injury. A1 competes with nitric oxide synthase (NOS) for their common substrate L-arginine. A1-mediated L-arginine depletion reduces nitric oxide (NO) formation by NOS leading to vascular dysfunction when endothelial NOS is involved but prevents inflammatory injury when inducible NOS is involved. Studies were performed using wild-type (WT) mice, global A1+/- knockout (KO), endothelial-specific A1 KO, and myeloid-specific A1 KO mice subjected to retinal IR injury. Global as well as myeloid-specific A1 KO mice showed worsened IR-induced neuronal loss and retinal thinning. Deletion of A1 in endothelial cells had no effect, while treatment with PEGylated (PEG) A1 improved neuronal survival in WT mice. In addition, A1+/- KO mice showed worsened vascular injury manifested by increased acellular capillaries. Western blotting analysis of retinal tissue showed increased inflammatory and necroptotic markers with A1 deletion. In vitro experiments showed that macrophages lacking A1 exhibit increased inflammatory response upon LPS stimulation. PEG-A1 treatment dampened this inflammatory response and decreased the LPS-induced metabolic reprogramming. Moreover, intravitreal injection of A1 KO macrophages or systemic macrophage depletion with clodronate liposomes increased neuronal loss after IR injury. These results demonstrate that A1 reduces IR injury-induced retinal neurovascular degeneration via dampening macrophage inflammatory responses. Increasing A1 offers a novel strategy for limiting neurovascular injury and promoting macrophage-mediated repair.


Subject(s)
Arginase/metabolism , Inflammation/metabolism , Ischemia/metabolism , Macrophages/metabolism , Reperfusion Injury/metabolism , Retina/metabolism , Retinal Neovascularization/metabolism , Animals , Apoptosis/physiology , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Retinal Diseases/metabolism
6.
Prog Retin Eye Res ; 67: 130-149, 2018 11.
Article in English | MEDLINE | ID: mdl-30075336

ABSTRACT

Retinal degenerative diseases are major causes of untreatable blindness worldwide and efficacious treatments for these diseases are sorely needed. A novel target for treatment of retinal disease is the transmembrane protein Sigma 1 Receptor (Sig1R). This enigmatic protein is an evolutionary isolate with no known homology to any other protein. Sig1R was originally thought to be an opioid receptor. That notion has been dispelled and more recent pharmacological and molecular studies suggest that it is a pluripotent modulator with a number of biological functions, many of which are relevant to retinal disease. This review provides an overview of the discovery of Sig1R and early pharmacologic studies that led to the cloning of the Sig1R gene and eventual elucidation of its crystal structure. Studies of Sig1R in the eye were not reported until the late 1990s, but since that time there has been increasing interest in the potential role of Sig1R as a target for retinal disease. Studies have focused on elucidating the mechanism(s) of Sig1R function in retina including calcium regulation, modulation of oxidative stress, ion channel regulation and molecular chaperone activity. Mechanistic studies have been performed in isolated retinal cells, such as Müller glial cells, microglial cells, optic nerve head astrocytes and retinal ganglion cells as well as in the intact retina. Several compelling studies have provided evidence of powerful in vivo neuroprotective effects against ganglion cell loss as well as photoreceptor cell loss. Also described are studies that have examined retinal structure/function in various models of retinal disease in which Sig1R is absent and reveal that these phenotypes are accelerated compared to retinas of animals that express Sig1R. The collective evidence from analysis of studies over the past 20 years is that Sig1R plays a key role in modulating retinal cellular stress and that it holds great promise as a target in retinal neurodegenerative disease.


Subject(s)
Molecular Targeted Therapy/methods , Neuroprotective Agents/therapeutic use , Receptors, sigma/physiology , Retinal Degeneration , Animals , Humans , Ion Channels/physiology , Molecular Chaperones/physiology , Neuroprotection/physiology , Oxidative Stress/physiology , Retinal Degeneration/drug therapy , Retinal Degeneration/metabolism , Sigma-1 Receptor
7.
Invest Ophthalmol Vis Sci ; 58(11): 4545-4558, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28877319

ABSTRACT

Purpose: Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods: Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results: Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 µV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions: Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.


Subject(s)
Apoptosis , Disease Models, Animal , Receptors, sigma/physiology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Animals , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Retina/physiology , Tomography, Optical Coherence , Sigma-1 Receptor
8.
Invest Ophthalmol Vis Sci ; 58(4): 1954-1963, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28384716

ABSTRACT

Purpose: Hyperhomocysteinemia (Hhcy) is implicated in certain retinal neurovascular diseases, although whether it is causative remains uncertain. In isolated ganglion cells (GCs), mild Hhcy induces profound death, whereas retinal phenotypes in Hhcy mice caused by mutations in remethylation (methylene tetrahydrofolatereductase [Mthfr+/-]) or transsulfuration pathways (cystathionine ß-synthase [Cbs+/-]) demonstrate mild GC loss and mild vasculopathy. The current work investigated compensation in vivo of one pathway for the other, and, because the transsulfuration pathway yields cysteine necessary for formation of glutathione (GSH), taurine, and hydrogen sulfide (H2S), they were analyzed also. Methods: Retinas isolated from wild-type (WT), Mthfr+/-, and Cbs+/- mice (12 and 22 weeks) were analyzed for methylene tetrahydrofolate reductase (MTHFR), cystathionine-ß-synthase (CBS), and cystathionase (CTH) RNA/protein levels. Retinas were evaluated for levels of reduced:oxidized GSH (GSH:GSSG), Slc7a11 (xCT), taurine, taurine transporter (TAUT), and H2S. Results: Aside from decreased CBS RNA/protein levels in Cbs+/- retinas, there were minimal alterations in remethylation/transsulfuration pathways in the two mutant mice strains. Glutathione and taurine levels in Mthfr+/- and Cbs+/- retinas were similar to WT, which may be due to robust levels of xCT and TAUT in mutant retinas. Interestingly, levels of H2S were markedly increased in retinas of Mthfr+/- and Cbs+/- mice compared with WT. Conclusions: Ganglion cell loss and vasculopathy observed in Mthfr+/- and Cbs+/- mouse retinas may be milder than expected, not because of compensatory increases of enzymes in remethylation/transsulfuration pathways, but because downstream transsulfuration pathway products GSH, taurine, and H2S are maintained at robust levels. Elevation of H2S is particularly intriguing owing to neuroprotective properties reported for this gasotransmitter.


Subject(s)
Gene Expression Regulation , Glutathione/metabolism , Hydrogen Sulfide/metabolism , Hyperhomocysteinemia/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Retinal Ganglion Cells/metabolism , Taurine/metabolism , Animals , Disease Models, Animal , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA/genetics , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retina/pathology , Retinal Diseases/etiology , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Ganglion Cells/pathology
9.
Doc Ophthalmol ; 134(3): 195-203, 2017 06.
Article in English | MEDLINE | ID: mdl-28349282

ABSTRACT

PURPOSE: Previous work has suggested that the retinal degeneration mutant rd8 mouse lacks an electroretinographic (ERG) phenotype until about 9 months of age. We evaluated the ERG phenotype of these mice by measuring both conventional ERG responses and scotopic threshold responses. METHODS: Groups of 4-month-old wild-type (WT) and mutant (rd8) mice were anesthetized and tested for mass retinal responses (ERGs) to several types of visual stimuli. Scotopic threshold responses were accumulated with brief scotopic flashes at a series of very dim intensities. Dark-adapted (scotopic) and light-adapted (photopic) responses to brief flashes at a series of higher intensities were recorded, along with long flashes and random modulations of light levels under photopic conditions. RESULTS: Negative scotopic threshold responses (nSTRs) had lower amplitudes in rd8 mice compared to WTs. Positive scotopic threshold responses were similar in the two groups. With the more intense stimuli, a- and c-wave amplitudes were smaller in rd8 mice. Both scotopic and photopic b-wave amplitudes tended to be larger in rd8 mice, though generally not significantly. CONCLUSIONS: The striking decrease in nSTR amplitudes was surprising, given that the main retinal effects of the rd8 mutation occur in the outer retina, at the external limiting membrane. The primary source of nSTRs in mice is thought to be at the amacrine cell level in the inner retina. Investigation of how this mutation leads to inner retinal dysfunction might reveal unexpected aspects of retinal cell biology and circuitry.


Subject(s)
Electroretinography , Retina/physiopathology , Retinal Degeneration/physiopathology , Animals , Color Vision/physiology , Dark Adaptation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Sensory Thresholds/physiology
10.
Adv Exp Med Biol ; 964: 267-284, 2017.
Article in English | MEDLINE | ID: mdl-28315277

ABSTRACT

This review article focuses on studies of Sigma 1 Receptor (Sigma1R) and retina . It provides a brief overview of the earliest pharmacological studies performed in the late 1990s that provided evidence of the presence of Sigma1R in various ocular tissues. It then describes work from a number of labs concerning the location of Sigma1R in several retinal cell types including ganglion, Müller glia , and photoreceptors . The role of Sigma1R ligands in retinal neuroprotection is emphasized. Early studies performed in vitro clearly showed that targeting Sigma1R could attenuate stress-induced retinal cell loss. These studies were followed by in vivo experiments. Data about the usefulness of targeting Sigma1R to prevent ganglion cell loss associated with diabetic retinopathy are reviewed. Mechanisms of Sigma1R-mediated retinal neuroprotection involving Müller cells , especially in modulating oxidative stress are described along with information about the retinal phenotype of mice lacking Sigma1R (Sigma1R -/- mice). The retina develops normally in Sigma1R -/- mice, but after many months there is evidence of apoptosis in the optic nerve head, decreased ganglion cell function and eventual loss of these cells. Additional studies using the Sigma1R -/- mice provide strong evidence that in the retina, Sigma1R plays a key role in modulating cellular stress. Recent work has shown that targeting Sigma1R may extend beyond protection of ganglion cells to include photoreceptor cell degeneration as well.


Subject(s)
Receptors, sigma/metabolism , Retina/metabolism , Animals , Ependymoglial Cells/metabolism , Humans , Oxidative Stress/physiology , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Sigma-1 Receptor
11.
Arch Virol ; 161(10): 2871-7, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27438076

ABSTRACT

Avian leukosis virus (ALV) is an avian oncogenic retrovirus that can induce various clinical tumors. The capsid protein P27 is the group-specific antigen of ALV and has many viral antigen sites that are easy to detect. In this study, we produced a monoclonal antibody (mAb), 3A9, that is specific for the P27 protein. A series of partially overlapping peptides were screened to define (181)PPSAR(185) as the minimal linear epitope recognized by mAb 3A9. The identified epitope could be recognized by chicken anti-ALV and mouse anti-ALV P27 sera. The epitope was highly conserved among a number of ALV-A, ALV-B and ALV-J strains. MAb 3A9 might be a valuable tool for the development of new immunodiagnostic approaches for ALV, and the defined linear epitope might help further our understanding of the antigenic structure of the P27 protein.


Subject(s)
Avian Leukosis Virus/immunology , Capsid Proteins/immunology , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Avian Leukosis Virus/genetics , Capsid Proteins/genetics , Chickens , Conserved Sequence , Epitopes, B-Lymphocyte/genetics , Female , Mice, Inbred BALB C
12.
Invest Ophthalmol Vis Sci ; 57(6): 2770-81, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27206247

ABSTRACT

PURPOSE: Sigma receptor 1 (Sigma1R), a nonopioid putative molecular chaperone, has neuroprotective properties in retina. This study sought to determine whether delaying administration of (+)-pentazocine, a high-affinity Sigma1R ligand after onset of diabetes in Ins2Akita/+ diabetic mice would afford retinal neuroprotection and to determine consequences on retinal phenotype in Ins2Akita/+ diabetic mice in the absence of Sigma1R. METHODS: Ins2Akita/+ diabetic and WT mice received intraperitoneal injections of (+)-pentazocine beginning 4 or 8 weeks after onset of diabetes; eyes were harvested at 25 weeks. Retinal histologic sections were analyzed to determine thicknesses of retinal layers, number of ganglion cells, and evidence of gliosis (increased glial fibrillary acidic protein levels). Ins2Akita/+/Sig1R-/-mice were generated and subjected to in vivo assessment of retinal architecture (optical coherence tomography [OCT]) and retinal vasculature using fluorescein angiography (FA) at 12 and 16 weeks compared with age-matched Ins2Akita/+ mice. Eyes were then harvested for retinal morphometric assessment and gliosis assessment. RESULTS: Wild-type mice had 13 ± 0.06 cells/100 µm retinal length; cell bodies in Ins2Akita/+ mice injected 4 and 8 weeks after onset of diabetes with (+)-pentazocine retained significantly more ganglion cells compared with Ins2Akita/+ mice (9 ± 0.04) and demonstrated significant attenuation of gliosis. Ins2Akita/+/Sig1R-/-mouse retinas, analyzed to determine whether the Ins2Akita/+ phenotype was accelerated when lacking Sigma1R, revealed increased nerve fiber layer thickness (OCT), evidence of vitreal opacities, and vessel beading (FA) compared with Ins2Akita/+ mice. Morphometric analysis revealed significantly fewer ganglion cells in Ins2Akita/+/Sig1R-/-mice compared with Ins2Akita/+ mice. CONCLUSIONS: Sigma1R may be a novel retinal stress modulator, and targeting it even after disease onset may afford retinal neuroprotection.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy/complications , Gene Expression Regulation , RNA/genetics , Receptors, sigma/genetics , Retinal Degeneration/etiology , Retinal Ganglion Cells/metabolism , Animals , Cell Death , Diabetic Retinopathy/metabolism , Female , Fluorescein Angiography , Fundus Oculi , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Receptors, sigma/biosynthesis , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Ganglion Cells/pathology , Tomography, Optical Coherence , Sigma-1 Receptor
13.
Exp Eye Res ; 145: 164-172, 2016 04.
Article in English | MEDLINE | ID: mdl-26646559

ABSTRACT

Mutations in crumb homologue 1 (CRB1) in humans are associated with Leber's congenital amaurosis (LCA) and retinitis pigmentosa (RP). There is no clear genotype-phenotype correlation for human CRB1 mutations in RP and LCA. The high variability in clinical features observed in CRB1 mutations suggests that environmental factors or genetic modifiers influence severity of CRB1 related retinopathies. Retinal degeneration 8 (rd8) is a spontaneous mutation in the Crb1 gene (Crb1(rdr/rd8)). Crb1(rdr/rd8) mice present with focal disruption in the outer retina manifesting as white spots on fundus examination. Mild retinal dysfunction with decreased b-wave amplitude has been reported in Crb1(rdr/rd8) mice at 18 months. Methylene tetrahydrofolate reductase (MTHFR) is a crucial enzyme of homocysteine metabolism. MTHFR mutations are prevalent in humans and are linked to a broad spectrum of disorders including cardiovascular and neurodegenerative diseases. We recently reported the retinal phenotype in Mthfr-deficient (Mthfr(+/-)) heterozygous mice. At 24 weeks the mice showed decreased RGC function, thinner nerve fiber layer, focal areas of vascular leakage and 20% fewer cells in the ganglion cell layer (GCL). Considering the variability in CRB1-related retinopathies and the high occurrence of human MTHFR mutations we evaluated whether Mthfr deficiency influences rd8 retinal phenotype. Mthfr heterozygous mice with rd8 mutations (Mthfr(+/-)(rd8/rd8)) and Crb(rd8/rd8) mice (Mthfr(+/+rd8/rd8)) mice were subjected to comprehensive retinal evaluation using ERG, fundoscopy, fluorescein angiography (FA), morphometric and retinal flat mount immunostaining analyses of isolectin-B4 at 8-54 wks. Assessment of retinal function revealed a significant decrease in the a-, b- and c-wave amplitudes in Mthfr(+/-)(rd8/rd8) mice at 52 wks. Fundoscopic evaluation demonstrated the presence of signature rd8 spots in Mthfr(+/+rd8/rd8) mice and an increase in the extent of these rd8 spots in Mthfr(+/-)(rd8/rd8) mice at 24 weeks and beyond. FA revealed marked vascular leakage, ischemia and vascular tortuosity in Mthfr(+/-)(rd8/rd8) mice at 24 and 52 weeks. Retinal dysplasia was observed in ∼14-33% Mthfr(+/-)(rd8/rd8) mice by morphometric analysis. This was accompanied by a ∼20% reduction in cells of the GCL of Mthfr(+/-)(rd8/rd8) mice at 24 and 52 weeks. Retinal flat mount immunostaining with isolectin-B4 showed neovascularization and loss of blood vessel integrity in Mthfr(+/-)(rd8/rd8) mice in contrast to mild vasculopathy in Mthfr(+/+rd8/rd8) mice. Taken together, our data support an earlier onset and worsened retinal phenotype when Mthfr and rd8 mutations coexist. Our study sets the stage for future studies to investigate the role of MTHFR deficiency in human CRB1 retinopathies.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Nerve Tissue Proteins/genetics , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Animals , DNA/genetics , DNA Mutational Analysis , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Genetic Association Studies , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology
14.
PLoS One ; 9(10): e109128, 2014.
Article in English | MEDLINE | ID: mdl-25343256

ABSTRACT

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a myocardial disease characterized by fibro-fatty replacement of myocardium in the right ventricular free wall and frequently results in life-threatening ventricular arrhythmias and sudden cardiac death. A heterozygous missense mutation in the transmembrane protein 43 (TMEM43) gene, p.S358L, has been genetically identified to cause autosomal dominant ARVC type 5 in a founder population from the island of Newfoundland, Canada. Little is known about the function of the TMEM43 protein or how it leads to the pathogenesis of ARVC. We sought to determine the distribution of TMEM43 and the effect of the p.S358L mutation on the expression and distribution of various intercalated (IC) disc proteins as well as functional effects on IC disc gap junction dye transfer and conduction velocity in cell culture. Through Western blot analysis, transmission electron microscopy (TEM), immunofluorescence (IF), and electrophysiological analysis, our results showed that the stable expression of p.S358L mutation in the HL-1 cardiac cell line resulted in decreased Zonula Occludens (ZO-1) expression and the loss of ZO-1 localization to cell-cell junctions. Junctional Plakoglobin (JUP) and α-catenin proteins were redistributed to the cytoplasm with decreased localization to cell-cell junctions. Connexin-43 (Cx43) phosphorylation was altered, and there was reduced gap junction dye transfer and conduction velocity in mutant TMEM43-transfected cells. These observations suggest that expression of the p.S358L mutant of TMEM43 found in ARVC type 5 may affect localization of proteins involved in conduction, alter gap junction function and reduce conduction velocity in cardiac tissue.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Endoplasmic Reticulum/genetics , Membrane Proteins/biosynthesis , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Cytoplasm , Desmosomes/metabolism , Gap Junctions/genetics , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Mutation, Missense , Myocardium/metabolism , Myocardium/pathology , Phosphorylation
15.
PLoS One ; 8(9): e72668, 2013.
Article in English | MEDLINE | ID: mdl-24039792

ABSTRACT

BACKGROUND: Congenital heart block (CHB) is a transplacentally acquired autoimmune disease associated with anti-Ro/SSA and anti-La/SSB maternal autoantibodies and is characterized primarily by atrioventricular (AV) block of the fetal heart. This study aims to investigate whether the T-type calcium channel subunit α1G may be a fetal target of maternal sera autoantibodies in CHB. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate differential mRNA expression of the T-type calcium channel CACNA1G (α1G gene) in the AV junction of human fetal hearts compared to the apex (18-22.6 weeks gestation). Using human fetal hearts (20-22 wks gestation), our immunoprecipitation (IP), Western blot analysis and immunofluorescence (IF) staining results, taken together, demonstrate accessibility of the α1G epitope on the surfaces of cardiomyocytes as well as reactivity of maternal serum from CHB affected pregnancies to the α1G protein. By ELISA we demonstrated maternal sera reactivity to α1G was significantly higher in CHB maternal sera compared to controls, and reactivity was epitope mapped to a peptide designated as p305 (corresponding to aa305-319 of the extracellular loop linking transmembrane segments S5-S6 in α1G repeat I). Maternal sera from CHB affected pregnancies also reacted more weakly to the homologous region (7/15 amino acids conserved) of the α1H channel. Electrophysiology experiments with single-cell patch-clamp also demonstrated effects of CHB maternal sera on T-type current in mouse sinoatrial node (SAN) cells. CONCLUSIONS/SIGNIFICANCE: Taken together, these results indicate that CHB maternal sera antibodies readily target an extracellular epitope of α1G T-type calcium channels in human fetal cardiomyocytes. CHB maternal sera also show reactivity for α1H suggesting that autoantibodies can target multiple fetal targets.


Subject(s)
Autoantibodies/immunology , Calcium Channels, T-Type/immunology , Epitopes/immunology , Heart Block/congenital , Amino Acid Sequence , Animals , Atrioventricular Node/drug effects , Atrioventricular Node/metabolism , Autoantibodies/blood , Autoantigens/immunology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Epitope Mapping , Extracellular Space , Female , Fetal Heart/drug effects , Fetal Heart/immunology , Fetal Heart/metabolism , Gene Expression , Heart Block/genetics , Heart Block/immunology , Humans , Male , Maternal-Fetal Exchange/immunology , Mice , Molecular Sequence Data , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Peptides/immunology , Pregnancy , Rabbits
16.
Brain Behav ; 1(2): 73-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22399087

ABSTRACT

Mutations in the L1 gene cause severe brain malformations and mental retardation. We investigated the potential roles of L1 in the regulation of choline acetyltransferase (ChAT) and in the development of septal cholinergic neurons, which are known to project to the hippocampus and play key roles in cognitive functions. Using stereological approaches, we detected significantly fewer ChAT-positive cholinergic neurons in the medial septum and vertical limb of the diagonal band of Broca (MS/VDB) of 2-week-old L1-deficient mice compared to wild-type littermates (1644 ± 137 vs. 2051 ± 165, P = 0.038). ChAT protein levels in the septum were 53% lower in 2-week-old L1-deficient mice compared to wild-type littermates. ChAT activity in the septum was significantly reduced in L1-deficient mice compared to wild-type littermates at 1 (34%) and 2 (40%) weeks of age. In vitro, increasing doses of L1-Fc induced ChAT activity in septal neurons with a significant linear trend (*P = 0.0065). At 4 weeks of age in the septum and at all time points investigated in the caudate-putamen (CPu), the number of ChAT-positive neurons and the levels of ChAT activity were not statistically different between L1-deficient mice and wild-type littermates. The total number of cells positive for the neuronal nuclear antigen (NeuN) in the MS/VDB and CPu was not statistically different in L1-deficient mice compared to wild-type littermates, and comparable expression of the cell cycle marker Ki67 was observed. Our results indicate that L1 is required for the timely maturation of septal cholinergic neurons and that L1 promotes the expression and activity of ChAT in septal neurons.

17.
J Cell Mol Med ; 13(10): 4219-28, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18662195

ABSTRACT

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a disorder of cardiomyocyte intercalated disk proteins causing sudden death. Heterozygous mutations of the desmosomal protein plakophilin-2 (PKP-2) are the commonest genetic cause of ARVC. Abnormal gap junction connexin43 expression has been reported in autosomal dominant forms of ARVC (Naxos and Carvajal disease) caused by homozygous mutations of desmosomal plakoglobin and desmoplakin. In tissue culture, suppression of PKP-2 results in decreased expression of connexin43. We sought to characterize the expression and localization of connexin43 in patients with ARVC secondary to heterozygous PKP-2 mutations. Complete PKP-2 gene sequencing of 27 ARVC patients was utilized to identify mutant genotypes. Endomyocardial biopsies of identified carriers were then assessed by immunofluorescence to visualize intercalated disk proteins. N-cadherin was targeted to highlight intercalated disks, followed by counterstaining for PKP-2 or connexin43 using confocal double immunofluorescence microscopy. Immunofluorescence was quantified using an AdobeA Photoshop protocol, and colocalization coefficients were determined. PKP-2 siRNA experiments were performed in mouse cardiomyocyte (HL1) cell culture with Western blot analysis to assess connexin43 expression following PKP-2 suppression. Missense and frameshift mutations of the PKP-2 gene were found in four patients with biopsy material available for analysis. Immunofluorescent studies showed PKP-2 localization to the intercalated disk despite mutations, but associated with decreased connexin43 expression and abnormal colocalization. PKP-2 siRNA in HL1 culture confirmed decreased connexin43 expression. Reduced connexin43 expression and localization to the intercalated disk occurs in heterozygous human PKP-2 mutations, potentially explaining the delayed conduction and propensity to develop arrhythmias seen in this disease.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Connexin 43/metabolism , Mutation/genetics , Plakophilins/genetics , Adolescent , Animals , Child, Preschool , Female , Fluorescent Antibody Technique , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Luminescent Measurements , Male , Mice , Microscopy, Confocal , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Plakophilins/chemistry , Plakophilins/metabolism , Protein Transport , RNA, Small Interfering/metabolism
18.
Brain Res ; 1144: 52-8, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17335782

ABSTRACT

Polysialic acid (PSA) is a large carbohydrate found exclusively on the neural cell adhesion molecule (NCAM). In the adult brain, PSA is re-expressed by septal axons sprouting and regenerating in an environment rich in laminin. Using an in vitro model, we tested the possibility that PSA limits septal outgrowth by preventing maximal interactions with a laminin substrate. Our results indicate that PSA removal from primary septal neurons plated on laminin significantly increased neurite outgrowth at 12 h (14%, p<0.05) and 24 h (22%, p<0.01). In contrast, the removal of PSA had no impact on septal neurite outgrowth on poly-D-lysine. PSA did not influence the plating adhesion of septal neurons on laminin or poly-D-lysine, indicating that the increase in neurite outgrowth caused by PSA removal on laminin is not related to the initial attachment of the neurons to this substrate. Neurite length on laminin was significantly reduced by the function-blocking beta1-integrin antibody in the presence of PSA (20% decrease, p<0.05), and following PSA removal (34% decrease compared to neurites treated with endoN and without the beta1-integrin antibody, p<0.01). Importantly, the beta1-integrin antibody completely abolished the neurite outgrowth promoting effect of PSA removal on laminin. The beta1-integrin antibody had no impact on septal neurite length on poly-D-lysine. Taken together, these results indicate that the removal of PSA from septal neurons increases neurite outgrowth on laminin by promoting interactions between beta1-integrin and laminin.


Subject(s)
Laminin/physiology , Neurites/drug effects , Neurons/cytology , Septum of Brain/cytology , Sialic Acids/pharmacology , Analysis of Variance , Animals , Antibodies/pharmacology , Drug Interactions , Embryo, Mammalian , GAP-43 Protein/metabolism , Laminin/immunology , Rats , Rats, Inbred F344 , Time Factors
19.
J Chromatogr A ; 1051(1-2): 147-53, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15532567

ABSTRACT

We demonstrate a convenient single-step quantitation technique for double-stranded DNA (dsDNA) fragments in polymerase chain reaction (PCR) products based on microchip capillary electrophoresis (micro-CE)/UV or fluorescence detection. PCR products of polymorphisms on the human Y-chromosome related to spermatogenic failure did not need purification. They were premixed and comigrated with a DNA digest whose concentration was known. Hydroxyethyl cellulose (HEC) dissolved in 5x Tris-borate-EDTA (5x TBE, pH 8.3) was used as a separation matrix in a linear polyacrylamide-coated quartz microchip, while mixed poly(ethyl oxides) (PEOs) of different molar-masses dissolved in 1 x TBE (pH 8.3) containing 1 ng/microl ethidium bromide was used as a separation matrix in an uncoated poly(methyl methacrylate) (PMMA) microchip. Elution profiles were monitored under either real-time linear imaging UV detection in the snapshot mode where the total separation time is fixed, or light-emitting diode (LED) confocal fluorescence detection in the finishline mode where solutes migrate over the same separation length. It is found that, in both modes, a linear relation exists between the peak areas (A) and the multiplication of the digest concentrations (C) and the fragment sizes (L) in a DNA restrictive digest. Using the comigration electropherogram of a single-step experiment, the concentrations of PCR products were directly determined using the A versus LC linear relationship. The sole condition to obey is that the chosen digest has different fragment sizes with the PCR products of interest. This condition is easy to obey, because micro-CE owns high separation ability, and many digests are commercially available. The recovery of the technique was between 98 and 105%. The R.S.D. for chip-to-chip concentration measurements was less than 6.0% (n = 6). Hence, the technique was accurate and reliable for DNA assays.


Subject(s)
DNA/analysis , Electrophoresis, Capillary/methods , Microchip Analytical Procedures , Spectrometry, Fluorescence/methods , Spectrophotometry, Ultraviolet/methods
20.
Life Sci ; 74(7): 897-907, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14659978

ABSTRACT

Oxidative stress plays a causative role in the development of hepatic fibrosis and apoptosis. Estradiol (E2) is an antioxidant, and idoxifene is a tissue-specific selective estrogen receptor modulator. We have previously demonstrated that E2 inhibits hepatic fibrosis in a rat model of hepatic fibrosis induced with dimethylnitrosamine (DMN), and suppresses activation of the nuclear factor (NF)-kappaB proinflammatory transcription factor in cultured rat hepatocytes undergoing oxidative stress. This study reports on the antioxidant and antiapoptotic role of idoxifene and E2 in the DMN model of hepatic fibrosis. The DMN model rats were administered with idoxifene or E2, and were examined activity of superoxide dismutase (SOD) and glutathione peroxidase (GPx) and expression of Bcl-2 family proteins in the liver. During the course of hepatofibrogenesis after DMN treatment, serum levels of lactate dehydrogenase (LDH), a biomarker for necrosis, and hepatic levels of malondialdehyde (MDA), an end product of lipid peroxidation, increased rapidly for 3 days. On day 14, serum LDH levels normalized, and hepatic fibrosis developed with increased levels of MDA and collagen and decreased production of SOD and GPx in the liver. Fibrotic liver also showed downregulation of Bcl-2 and Bcl-X(L) expression and upregulation of Bad expression. Idoxifene and E2 suppressed DMN-mediated necrosis, lipid peroxidation, the loss of antioxidant enzyme activity, and proapoptotic status in Bcl-2 family protein expression as well as hepatic fibrosis. These findings indicate that, in addition to their antiinflammatory and antifibrotic action, idoxifene and E2 could enhance antioxidant and antiapoptotic activity in hepatic fibrosis in rats.


Subject(s)
Antioxidants/therapeutic use , Apoptosis/drug effects , Estradiol/therapeutic use , Estrogen Antagonists/therapeutic use , Liver Cirrhosis, Experimental/drug therapy , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Administration, Oral , Animals , Carrier Proteins/metabolism , Dimethylnitrosamine/toxicity , Disease Models, Animal , Drug Therapy, Combination , Estradiol/administration & dosage , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , Glutathione Peroxidase/metabolism , Injections, Intraperitoneal , L-Lactate Dehydrogenase/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/pathology , Male , Malondialdehyde/blood , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Tamoxifen/administration & dosage , bcl-Associated Death Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...