Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Microvasc Res ; 151: 104609, 2024 01.
Article in English | MEDLINE | ID: mdl-37716411

ABSTRACT

OBJECTIVE: Vascular smooth muscle cell (VSMC) phenotypic switching is critical for normal vessel formation, vascular stability, and healthy brain aging. Phenotypic switching is regulated by mediators including platelet derived growth factor (PDGF)-BB, insulin-like growth factor (IGF-1), as well as transforming growth factor-ß (TGF-ß) and endothelin-1 (ET-1), but much about the role of these factors in microvascular VSMCs remains unclear. METHODS: We used primary rat microvascular VSMCs to explore PDGF-BB- and IGF-1-induced phenotypic switching. RESULTS: PDGF-BB induced an early proliferative response, followed by formation of polarized leader cells and rapid, directionally coordinated migration. In contrast, IGF-1 induced cell hypertrophy, and only a small degree of migration by unpolarized cells. TGF-ß and ET-1 selectively inhibit PDGF-BB-induced VSMC migration primarily by repressing migratory polarization and formation of leader cells. Contractile genes were downregulated by both growth factors, while other genes were differentially regulated by PDGF-BB and IGF-1. CONCLUSIONS: These studies indicate that PDGF-BB and IGF-1 stimulate different types of microvascular VSMC phenotypic switching characterized by different modes of cell migration. Our studies are consistent with a chronic vasoprotective role for IGF-1 in VSMCs in the microvasculature while PDGF is more involved in VSMC proliferation and migration in response to acute activities such as neovascularization. Better understanding of the nuances of the phenotypic switching induced by these growth factors is important for our understanding of a variety of microvascular diseases.


Subject(s)
Insulin-Like Growth Factor I , Rats , Animals , Becaplermin/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Proto-Oncogene Proteins c-sis/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/metabolism , Transforming Growth Factor beta/metabolism , Myocytes, Smooth Muscle , Cell Proliferation , Cell Movement , Cells, Cultured
2.
Biomolecules ; 13(12)2023 11 23.
Article in English | MEDLINE | ID: mdl-38136563

ABSTRACT

The bioactive sphingolipid sphingosine-1-phosphate (S1P) acts as a ligand for a family of G protein-coupled S1P receptors (S1PR1-5) to participate in a variety of signaling pathways. However, their specific roles in the neural retina remain unclear. We previously showed that S1P receptor subtype 2 (S1PR2) is expressed in murine retinas, primarily in photoreceptors and bipolar cells, and its expression is altered by retinal stress. This study aims to elucidate the role of S1PR2 in the mouse retina. We examined light responses by electroretinography (ERG), structural differences by optical coherence tomography (OCT), and protein levels by immunohistochemistry (IHC) in wild-type (WT) and S1PR2 knockout (KO) mice at various ages between 3 and 6 months. We found that a- and b-wave responses significantly increased at flash intensities between 400~2000 and 4~2000 cd.s/m2, respectively, in S1PR2 KO mice relative to those of WT controls at baseline. S1PR2 KO mice also exhibited significantly increased retinal nerve fiber layer (RNFL) and outer plexiform layer (OPL) thickness by OCT relative to the WT. Finally, in S1PR2 KO mice, we observed differential labeling of synaptic markers by immunohistochemistry (IHC) and quantitative reverse transcription polymerase chain reaction (RT-qPCR). These results suggest a specific involvement of S1PR2 in the structure and synaptic organization of the retina and a potential role in light-mediated functioning of the retina.


Subject(s)
Electroretinography , Retina , Mice , Animals , Sphingosine-1-Phosphate Receptors/metabolism , Retina/metabolism , Signal Transduction , Mice, Knockout
3.
Cell Death Dis ; 14(7): 420, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443173

ABSTRACT

Retinitis pigmentosa (RP) defines a group of hereditary progressive rod-cone degenerations that exhibit a common phenotype caused by variants in over 70 genes. While most variants in the dehydrodolichyl diphosphate synthase (DHDDS) gene result in syndromic abnormalities, some variants cause non-syndromic RP (RP59). DHDDS encodes one subunit of the enzyme cis-prenyltransferase (CPT), which is required for the synthesis of dolichol (Dol), that is a necessary protein glycosylation cofactor. We previously reported the creation and initial characterization of a knock-in (KI) mouse model harboring the most prevalent RP59-associated DHDDS variant (K42E) to understand how defects in DHDDS lead to retina-specific pathology. This model exhibited no profound retinal degeneration, nor protein N-glycosylation defects. Here, we report that the Dol isoprenylogue species in retina, liver, and brain of the K42E mouse model are statistically shorter than in the corresponding tissues of age-matched controls, as reported in blood and urine of RP59 patients. Retinal transcriptome analysis demonstrated elevation of many genes encoding proteins involved in synaptogenesis and synaptic function. Quantitative retinal cell layer thickness measurements demonstrated a significant reduction in the inner nuclear layer (INL) and total retinal thickness (TRT) beginning at postnatal (PN) ∼2 months, progressively increasing to PN 18-mo. Histological analysis revealed cell loss in the INL, outer plexiform layer (OPL) disruption, and ectopic localization of outer nuclear layer (ONL) nuclei into the OPL of K42E mutant retinas, relative to controls. Electroretinograms (ERGs) of mutant mice exhibited reduced b-wave amplitudes beginning at PN 1-mo, progressively declining through PN 18-mo, without appreciable a-wave attenuation, relative to controls. Our results suggest that the underlying cause of DHDDS K42E variant driven RP59 retinal pathology is defective synaptic transmission from outer to inner retina.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Animals , Mice , Retina/metabolism , Retinal Degeneration/metabolism , Retinitis Pigmentosa/metabolism , Electroretinography , Synaptic Transmission
4.
J Neurosci ; 43(33): 5963-5974, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37491316

ABSTRACT

Elongation of very long fatty acids-4 (ELOVL4) mediates biosynthesis of very long chain-fatty acids (VLC-FA; ≥28 carbons). Various mutations in this enzyme result in spinocerebellar ataxia-34 (SCA34). We generated a rat model of human SCA34 by knock-in of a naturally occurring c.736T>G, p.W246G mutation in the Elovl4 gene. Our previous analysis of homozygous W246G mutant ELOVL4 rats (MUT) revealed early-onset gait disturbance and impaired synaptic transmission and plasticity at parallel fiber-Purkinje cell (PF-PC) and climbing fiber-Purkinje cell (CF-PC) synapses. However, the underlying mechanisms that caused these defects remained unknown. Here, we report detailed patch-clamp recordings from Purkinje cells that identify impaired synaptic mechanisms. Our results show that miniature EPSC (mEPSC) frequency is reduced in MUT rats with no change in mEPSC amplitude, suggesting a presynaptic defect of excitatory synaptic transmission on Purkinje cells. We also find alterations in inhibitory synaptic transmission as miniature IPSC (mIPSC) frequency and amplitude are increased in MUT Purkinje cells. Paired-pulse ratio is reduced at PF-PC synapses but increased at CF-PC synapses in MUT rats, which along with results from high-frequency stimulation suggest opposite changes in the release probability at these two synapses. In contrast, we identify exaggerated persistence of EPSC amplitude at CF-PC and PF-PC synapses in MUT cerebellum, suggesting a larger readily releasable pool (RRP) at both synapses. Furthermore, the dendritic spine density is reduced in MUT Purkinje cells. Thus, our results uncover novel mechanisms of action of VLC-FA at cerebellar synapses, and elucidate the synaptic dysfunction underlying SCA34 pathology.SIGNIFICANCE STATEMENT Very long chain-fatty acids (VLC-FA) are an understudied class of fatty acids that are present in the brain. They are critical for brain function as their deficiency caused by mutations in elongation of very long fatty acids-4 (ELOVL4), the enzyme that mediates their biosynthesis, results in neurologic diseases including spinocerebellar ataxia-34 (SCA34), neuroichthyosis, and Stargardt-like macular dystrophy. In this study, we investigated the synaptic defects present in a rat model of SCA34 and identified defects in presynaptic neurotransmitter release and dendritic spine density at synapses in the cerebellum, a brain region involved in motor coordination. These results advance our understanding of the synaptic mechanisms regulated by VLC-FA and describe the synaptic dysfunction that leads to motor incoordination in SCA34.


Subject(s)
Cerebellum , Spinocerebellar Ataxias , Rats , Humans , Animals , Cerebellum/physiology , Synapses/physiology , Synaptic Transmission/physiology , Ataxia/genetics , Purkinje Cells/physiology , Spinocerebellar Ataxias/genetics , Fatty Acids , Eye Proteins/metabolism , Membrane Proteins/metabolism
5.
MethodsX ; 9: 101796, 2022.
Article in English | MEDLINE | ID: mdl-36042811

ABSTRACT

Expansion microscopy (ExM) is a microscopic imaging approach that can achieve super-resolution visualization of fluorescently labeled biological samples using conventional fluorescence microscopy. The method is based on embedding of a fluorescently labeled biological sample in a hydrogel matrix followed by the physical expansion of the specimen, which is then viewed using a conventional fluorescent microscope. Variations of the method can be used to visualize endogenously expressed fluorescent proteins, such as GFP, fluorescently tagged antibodies, nucleic acids, or other fluorescently tagged molecules. A significant challenge of the method is that the physical expansion of the specimen produces a concommitant reduction in fluorescence intensity, which can make imaging difficult. We describe an approach for amplifying fluorescence signal following expansion of immunolabeled tissue sections by applying fluorescently labeled Fab fragment secondary antibodies to intensify fluorescent signal and enhance detection of labeling using conventional fluorescent microscopy. A method to increase immunofluorescence signal intensity of Expansion Microscopy specimens is described. Method utilizes commercially available reagents. Enhances ability to acquire useful images in expanded tissue samples.

6.
J Neurosci ; 42(31): 5992-6006, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35760531

ABSTRACT

Cognitive decline is a debilitating aspect of aging and neurodegenerative diseases such as Alzheimer's disease are closely associated with mitochondrial dysfunction, increased reactive oxygen species, neuroinflammation, and astrogliosis. This study investigated the effects of decreased mitochondrial antioxidant response specifically in astrocytes on cognitive performance and neuronal function in C57BL/6J mice using a tamoxifen-inducible astrocyte-specific knockout of manganese superoxide dismutase (aSOD2-KO), a mitochondrial matrix antioxidant that detoxifies superoxide generated during mitochondrial respiration. We reduced astrocyte SOD2 levels in male and female mice at 11-12 months of age and tested in an automated home cage (PhenoTyper) apparatus for diurnal patterns, spatial learning, and memory function at 15 months of age. aSOD2-KO impaired hippocampal-dependent spatial working memory and decreased cognitive flexibility in the reversal phase of the testing paradigm in males. Female aSOD2-KO showed no learning and memory deficits compared with age-matched controls despite significant reduction in hippocampal SOD2 expression. aSOD2-KO males further showed decreased hippocampal long-term potentiation, but paired-pulse facilitation was unaffected. Levels of d-serine, an NMDA receptor coagonist, were also reduced in aSOD2-KO mice, but female knockouts showed a compensatory increase in serine racemase expression. Furthermore, aSOD2-KO mice demonstrated increased density of astrocytes, indicative of astrogliosis, in the hippocampus compared with age-matched controls. These data demonstrate that reduction in mitochondrial antioxidant stress response in astrocytes recapitulates age-related deficits in cognitive function, d-serine availability, and astrogliosis. Therefore, improving astrocyte mitochondrial homeostasis may provide a therapeutic target for intervention for cognitive impairment in aging.SIGNIFICANCE STATEMENT Diminished antioxidant response is associated with increased astrogliosis in aging and in Alzheimer's disease. Manganese superoxide dismutase (SOD2) is an antioxidant in the mitochondrial matrix that detoxifies superoxide and maintains mitochondrial homeostasis. We show that astrocytic ablation of SOD2 impairs hippocampal-dependent plasticity in spatial working memory, reduces long-term potentiation of hippocampal neurons and levels of the neuromodulator d-serine, and increases astrogliosis, consistent with defects in advanced aging and Alzheimer's disease. Our data provide strong evidence for sex-specific effects of astrocytic SOD2 functions in age-related cognitive dysfunction.


Subject(s)
Alzheimer Disease , Astrocytes , Superoxide Dismutase , Alzheimer Disease/metabolism , Animals , Antioxidants/metabolism , Astrocytes/metabolism , Cognition/physiology , Female , Gliosis/metabolism , Hippocampus/metabolism , Male , Memory, Short-Term , Mice , Mice, Inbred C57BL , Serine/metabolism , Sex Factors , Superoxide Dismutase/genetics , Superoxides/metabolism
7.
Mol Neurobiol ; 58(10): 4921-4943, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34227061

ABSTRACT

Spinocerebellar ataxia (SCA) is a neurodegenerative disorder characterized by ataxia and cerebellar atrophy. A number of different mutations gives rise to different types of SCA with characteristic ages of onset, symptomatology, and rates of progression. SCA type 34 (SCA34) is caused by mutations in ELOVL4 (ELOngation of Very Long-chain fatty acids 4), a fatty acid elongase essential for biosynthesis of Very Long Chain Saturated and Polyunsaturated Fatty Acids (VLC-SFA and VLC-PUFA, resp., ≥28 carbons), which have important functions in the brain, skin, retina, Meibomian glands, testes, and sperm. We generated a rat model of SCA34 by knock-in of the SCA34-causing 736T>G (p.W246G) ELOVL4 mutation. Rats carrying the mutation developed impaired motor deficits by 2 months of age. To understand the mechanism of these motor deficits, we performed electrophysiological studies using cerebellar slices from rats homozygous for W246G mutant ELOVL4 and found marked reduction of long-term potentiation at parallel fiber synapses and long-term depression at climbing fiber synapses onto Purkinje cells. Neuroanatomical analysis of the cerebellum showed normal cytoarchitectural organization with no evidence of degeneration out to 6 months of age. These results point to ELOVL4 as essential for motor function and cerebellar synaptic plasticity. The results further suggest that ataxia in SCA34 patients may arise from a primary impairment of synaptic plasticity and cerebellar network desynchronization before onset of neurodegeneration and progression of the disease at a later age.


Subject(s)
Eye Proteins/genetics , Membrane Proteins/genetics , Mutation/genetics , Nerve Fibers, Myelinated/pathology , Neuronal Plasticity/physiology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Animals , Cerebellum/pathology , Female , Male , Motor Disorders/genetics , Motor Disorders/pathology , Organ Culture Techniques , Rats , Rats, Long-Evans , Rats, Transgenic
8.
Mol Neurobiol ; 57(11): 4735-4753, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32780351

ABSTRACT

Elongation of very long chain fatty acids-4 (ELOVL4) is essential for synthesis of very long chain polyunsaturated and saturated fatty acids (VLC-PUFA and VLC-SFA, respectively) of chain length greater than 26 carbons. Mutations in the ELOVL4 gene cause several distinct neurodegenerative diseases including Stargardt-like macular dystrophy (STGD3), spinocerebellar ataxia 34 (SCA34), and a neuro-ichthyotic syndrome with severe seizures and spasticity, as well as erythrokeratitis variabilis (EKV), a skin disorder. However, the relationship between ELOVL4 mutations, its VLC-PUFA and VLC-SFA products, and specific neurological symptoms remains unclear. We generated a knock-in rat line (SCA34-KI) that expresses the 736T>G (p.W246G) form of ELOVL4 that causes human SCA34. Lipids were analyzed by gas chromatography and mass spectrometry. Retinal function was assessed using electroretinography. Retinal integrity was assessed by histology, optical coherence tomography, and immunolabeling. Analysis of retina and skin lipids showed that the W246G mutation selectively impaired synthesis of VLC-SFA, but not VLC-PUFA. Homozygous SCA34-KI rats showed reduced ERG a- and b-wave amplitudes by 90 days of age, particularly for scotopic responses. Anatomical analyses revealed no indication of neurodegeneration in heterozygote or homozygote SCA34-KI rats out to 6-7 months of age. These studies reveal a previously unrecognized role for VLC-SFA in regulating retinal function, particularly transmission from photoreceptors to the inner retina, in the absence of neurodegeneration. Furthermore, these findings suggest that the tissue specificity and symptoms associated with disease-causing ELOVL4 mutations likely arise from selective differences in the ability of the mutant ELOVL4 enzymes to support synthesis of VLC-PUFA and/or VLC-SFA.


Subject(s)
Eye Proteins/genetics , Membrane Proteins/genetics , Mutation/genetics , Photoreceptor Cells, Vertebrate/pathology , Retina/physiopathology , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Animals , Disease Models, Animal , Electroretinography , Fatty Acids/metabolism , Humans , Night Vision , Phenotype , Rats , Rats, Transgenic
9.
Proc Natl Acad Sci U S A ; 117(34): 20615-20624, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32778589

ABSTRACT

Trafficking of photoreceptor membrane proteins from their site of synthesis in the inner segment (IS) to the outer segment (OS) is critical for photoreceptor function and vision. Here we evaluate the role of syntaxin 3 (STX3), in trafficking of OS membrane proteins such as peripherin 2 (PRPH2) and rhodopsin. Photoreceptor-specific Stx3 knockouts [Stx3f/f(iCre75) and Stx3f/f(CRX-Cre) ] exhibited rapid, early-onset photoreceptor degeneration and functional decline characterized by structural defects in IS, OS, and synaptic terminals. Critically, in the absence of STX3, OS proteins such as PRPH2, the PRPH2 binding partner, rod outer segment membrane protein 1 (ROM1), and rhodopsin were mislocalized along the microtubules to the IS, cell body, and synaptic region. We find that the PRPH2 C-terminal domain interacts with STX3 as well as other photoreceptor SNAREs, and our findings indicate that STX3 is an essential part of the trafficking pathway for both disc (rhodopsin) and rim (PRPH2/ROM1) components of the OS.


Subject(s)
Peripherins/metabolism , Qa-SNARE Proteins/metabolism , Retinal Photoreceptor Cell Inner Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Rhodopsin/metabolism , Animals , Gene Knockdown Techniques , Mice , Photoreceptor Cells, Vertebrate/physiology , Protein Transport , Qa-SNARE Proteins/genetics , Retinal Photoreceptor Cell Inner Segment/ultrastructure , Retinal Photoreceptor Cell Outer Segment/ultrastructure , SNARE Proteins/metabolism
10.
Front Cell Neurosci ; 13: 428, 2019.
Article in English | MEDLINE | ID: mdl-31616255

ABSTRACT

Elongation of Very Long chain fatty acids-4 (ELOVL4) protein is a member of the ELOVL family of fatty acid elongases that is collectively responsible for catalyzing formation of long chain fatty acids. ELOVL4 is the only family member that catalyzes production of Very Long Chain Saturated Fatty Acids (VLC-SFA) and Very Long Chain Polyunsaturated Fatty Acids (VLC-PUFA) with chain lengths ≥28 carbons. ELOVL4 and its VLC-SFA and VLC-PUFA products are emerging as important regulators of synaptic signaling and neuronal survival in the central nervous system (CNS). Distinct sets of mutations in ELOVL4 cause three different neurological diseases in humans. Heterozygous inheritance of one set of autosomal dominant ELOVL4 mutations that leads to truncation of the ELOVL4 protein causes Stargardt-like macular dystrophy (STGD3), an aggressive juvenile-onset retinal degeneration. Heterozygous inheritance of a different set of autosomal dominant ELOVL4 mutations that leads to a full-length protein with single amino acid substitutions causes spinocerebellar ataxia 34 (SCA34), a late-onset neurodegenerative disease characterized by gait ataxia and cerebellar atrophy. Homozygous inheritance of a different set of ELOVL4 mutations causes a more severe disease with infantile onset characterized by seizures, spasticity, intellectual disability, ichthyosis, and premature death. ELOVL4 is expressed widely in the CNS and is found primarily in neurons. ELOVL4 is expressed in cell-specific patterns within different regions of the CNS that are likely to be related to disease symptoms. In the retina, ELOVL4 is expressed exclusively in photoreceptors and produces VLC-PUFA that are incorporated into phosphatidylcholine and enriched in the light sensitive membrane disks of the photoreceptor outer segments. VLC-PUFA are enzymatically converted into "elovanoid" compounds that appear to provide paracrine signals that promote photoreceptor and neuronal survival. In the brain, the main ELOVL4 products are VLC-SFA that are incorporated into sphingolipids and enriched in synaptic vesicles, where they regulate kinetics of presynaptic neurotransmitter release. Understanding the function of ELOVL4 and its VLC-SFA and VLC-PUFA products will advance our understanding of basic mechanisms in neural signaling and has potential for developing novel therapies for seizure and neurodegenerative diseases.

11.
Behav Brain Res ; 340: 183-194, 2018 03 15.
Article in English | MEDLINE | ID: mdl-27793733

ABSTRACT

Mild traumatic brain injury (mTBI) diagnoses have increased due to aggressive sports and blast-related injuries, but the cellular mechanisms and pathology underlying mTBI are not completely understood. Previous reports indicate that Nociceptin Orphanin/FQ (N/OFQ), an endogenous neuropeptide, contributes to post-injury ischemia following mechanical brain injury, yet its specific role in cerebral hypoxia, vestibulomotor function and injury marker expression following blast-induced TBI is not known. This study is the first to identify a direct association of N/OFQ and its N/OFQ peptide (NOP) receptor with TBI-induced changes following a single 80psi head blast exposure in male rats. N/OFQ and NOP receptor expression increased in brain tissue and plasma following TBI, concurrent with vestibular dysfunction but preceding hypoxia and appearance of injury markers compared to sham rats. A single post-blast treatment with the NOP receptor antagonist, SB-612111, transiently improved acute vestibulomotor performance. It also prevented increases in markers of TBI-induced hypoxia, pro-apoptotic proteins and injury seen 8-10days post-blast. This study reveals an apparent role for the N/OFQ-NOP receptor system in blast TBI and suggests potential therapeutic utility of NOP receptor antagonists for mTBI.


Subject(s)
Blast Injuries/drug therapy , Brain Concussion/drug therapy , Brain/drug effects , Cycloheptanes/pharmacology , Hypoxia, Brain/prevention & control , Narcotic Antagonists/pharmacology , Piperidines/pharmacology , Animals , Blast Injuries/pathology , Blast Injuries/physiopathology , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Brain Concussion/etiology , Brain Concussion/pathology , Brain Concussion/physiopathology , Hypoxia, Brain/etiology , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Male , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Proteome/drug effects , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Nociceptin Receptor
12.
Mol Neurobiol ; 55(2): 1795-1813, 2018 02.
Article in English | MEDLINE | ID: mdl-29168048

ABSTRACT

Lipids are essential components of the nervous system. However, the functions of very long-chain fatty acids (VLC-FA; ≥ 28 carbons) in the brain are unknown. The enzyme ELOngation of Very Long-chain fatty acids-4 (ELOVL4) catalyzes the rate-limiting step in the biosynthesis of VLC-FA (Agbaga et al., Proc Natl Acad Sci USA 105(35): 12843-12848, 2008; Logan et al., J Lipid Res 55(4): 698-708, 2014), which we identified in the brain as saturated fatty acids (VLC-SFA). Homozygous mutations in ELOVL4 cause severe neuropathology in humans (Ozaki et al., JAMA Neurol 72(7): 797-805, 2015; Mir et al., BMC Med Genet 15: 25, 2014; Cadieux-Dion et al., JAMA Neurol 71(4): 470-475, 2014; Bourassa et al., JAMA Neurol 72(8): 942-943, 2015; Aldahmesh et al., Am J Hum Genet 89(6): 745-750, 2011) and are post-natal lethal in mice (Cameron et al., Int J Biol Sci 3(2): 111-119, 2007; Li et al., Int J Biol Sci 3(2): 120-128, 2007; McMahon et al., Molecular Vision 13: 258-272, 2007; Vasireddy et al., Hum Mol Genet 16(5): 471-482, 2007) from dehydration due to loss of VLC-SFA that comprise the skin permeability barrier. Double transgenic mice with homozygous knock-in of the Stargardt-like macular dystrophy (STDG3; 797-801_AACTT) mutation of Elovl4 with skin-specific rescue of wild-type Elovl4 expression (S + Elovl4 mut/mut mice) develop seizures by P19 and die by P21. Electrophysiological analyses of hippocampal slices showed aberrant epileptogenic activity in S + Elovl4 mut/mut mice. FM1-43 dye release studies showed that synapses made by cultured hippocampal neurons from S + Elovl4 mut/mut mice exhibited accelerated synaptic release kinetics. Supplementation of VLC-SFA to cultured hippocampal neurons from mutant mice rescued defective synaptic release to wild-type rates. Together, these studies establish a critical, novel role for ELOVL4 and its VLC-SFA products in regulating synaptic release kinetics and epileptogenesis. Future studies aimed at understanding the molecular mechanisms by which VLC-SFA regulate synaptic function may provide new targets for improved seizure therapies.


Subject(s)
Eye Proteins/metabolism , Fatty Acids/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Mutation , Seizures/metabolism , Animals , Disease Models, Animal , Eye Proteins/genetics , Fatty Acids/pharmacology , Hippocampus/drug effects , Macular Degeneration/genetics , Macular Degeneration/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Seizures/genetics
13.
Front Neuroanat ; 11: 38, 2017.
Article in English | MEDLINE | ID: mdl-28507511

ABSTRACT

ELOngation of Very Long chain fatty acids (ELOVL)-4 is essential for the synthesis of very long chain-fatty acids (fatty acids with chain lengths ≥ 28 carbons). The functions of ELOVL4 and its very long-chain fatty acid products are poorly understood at present. However, mutations in ELOVL4 cause neurodevelopmental or neurodegenerative diseases that vary according to the mutation and inheritance pattern. Heterozygous inheritance of different ELOVL4 mutations causes Stargardt-like Macular Dystrophy or Spinocerebellar Ataxia type 34. Homozygous inheritance of ELOVL4 mutations causes more severe disease characterized by seizures, intellectual disability, ichthyosis, and premature death. To better understand ELOVL4 and very long chain fatty acid function in the brain, we examined ELOVL4 expression in the mouse brain between embryonic day 18 and postnatal day 60 by immunolabeling using ELOVL4 and other marker antibodies. ELOVL4 was widely expressed in a region- and cell type-specific manner, and was restricted to cell bodies, consistent with its known localization to endoplasmic reticulum. ELOVL4 labeling was most prominent in gray matter, although labeling also was present in some cells located in white matter. ELOVL4 was widely expressed in the developing brain by embryonic day 18 and was especially pronounced in regions underlying the lateral ventricles and other neurogenic regions. The basal ganglia in particular showed intense ELOVL4 labeling at this stage. In the postnatal brain, cerebral cortex, hippocampus, cerebellum, thalamus, hypothalamus, midbrain, pons, and medulla all showed prominent ELOVL4 labeling, although ELOVL4 distribution was not uniform across all cells or subnuclei within these regions. In contrast, the basal ganglia showed little ELOVL4 labeling in the postnatal brain. Double labeling studies showed that ELOVL4 was primarily expressed by neurons, although presumptive oligodendrocytes located in white matter tracts also showed labeling. Little or no ELOVL4 labeling was present in astrocytes or radial glial cells. These findings suggest that ELOVL4 and its very long chain fatty acid products are important in many parts of the brain and that they are particularly associated with neuronal function. Specific roles for ELOVL4 and its products in oligodendrocytes and myelin and in cellular proliferation, especially during development, are possible.

14.
Diabetes ; 64(10): 3554-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26068541

ABSTRACT

To dissect the role of vascular endothelial growth factor receptor-2 (VEGFR2) in Müller cells and its effect on neuroprotection in diabetic retinopathy (DR), we disrupted VEGFR2 in mouse Müller glia and determined its effect on Müller cell survival, neuronal integrity, and trophic factor production in diabetic retinas. Diabetes was induced with streptozotocin. Retinal function was measured with electroretinography. Müller cell and neuronal densities were assessed with morphometric and immunohistochemical analyses. Loss of VEGFR2 caused a gradual reduction in Müller glial density, which reached to a significant level 10 months after the onset of diabetes. This observation was accompanied by an age-dependent decrease of scotopic and photopic electroretinography amplitudes and accelerated loss of rod and cone photoreceptors, ganglion cell layer cells, and inner nuclear layer neurons and by a significant reduction of retinal glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor. Our results suggest that VEGFR2-mediated Müller cell survival is required for the viability of retinal neurons in diabetes. The genetically altered mice established in this study can be used as a diabetic animal model of nontoxin-induced Müller cell ablation, which will be useful for exploring the cellular mechanisms of neuronal alteration in DR.


Subject(s)
Diabetic Retinopathy/pathology , Ependymoglial Cells/physiology , Retinal Neurons/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Electroretinography , Gene Expression Regulation , Mice , Mice, Knockout , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics
15.
J Neurosci ; 33(32): 13053-65, 13065a, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926259

ABSTRACT

Horizontal cells are interneurons that synapse with photoreceptors in the outer retina. Their genesis during development is subject to regulation by transcription factors in a hierarchical manner. Previously, we showed that Onecut 1 (Oc1), an atypical homeodomain transcription factor, is expressed in developing horizontal cells (HCs) and retinal ganglion cells (RGCs) in the mouse retina. Herein, by knocking out Oc1 specifically in the developing retina, we show that the majority (∼80%) of HCs fail to form during early retinal development, implying that Oc1 is essential for HC genesis. However, no other retinal cell types, including RGCs, were affected in the Oc1 knock-out. Analysis of the genetic relationship between Oc1 and other transcription factor genes required for HC development revealed that Oc1 functions downstream of FoxN4, in parallel with Ptf1a, but upstream of Lim1 and Prox1. By in utero electroporation, we found that Oc1 and Ptf1a together are not only essential, but also sufficient for determination of HC fate. In addition, the synaptic connections in the outer plexiform layer are defective in Oc1-null mice, and photoreceptors undergo age-dependent degeneration, indicating that HCs are not only an integral part of the retinal circuitry, but also are essential for the survival of photoreceptors. In sum, these results demonstrate that Oc1 is a critical determinant of HC fate, and reveal that HCs are essential for photoreceptor viability, retinal integrity, and normal visual function.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hepatocyte Nuclear Factor 6/metabolism , Neurogenesis/genetics , Retina/cytology , Retinal Horizontal Cells/metabolism , Animals , Cell Count , Cell Differentiation/genetics , Cell Survival , Embryo, Mammalian , Eye Proteins/genetics , Green Fluorescent Proteins/genetics , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Neuroglia/metabolism , Neuroglia/physiology , Neurons/classification , Neurons/metabolism , Neurons/ultrastructure , Protein Kinase C-alpha/metabolism , Retina/embryology , Retinal Horizontal Cells/ultrastructure , Synapses/metabolism , Synapses/ultrastructure , Transcription Factors/genetics , Transcription Factors/metabolism , Homeobox Protein SIX3
16.
PLoS One ; 8(6): e67381, 2013.
Article in English | MEDLINE | ID: mdl-23840687

ABSTRACT

Retinal cells become post-mitotic early during post-natal development. It is likely that p53, a well-known cell cycle regulator, is involved in regulating the genesis, differentiation and death of retinal cells. Furthermore, retinal cells are under constant oxidative stress that can result in DNA damage, due to the extremely high level of metabolic activity associated with phototransduction. If not repaired, this damage may result in p53-dependent cell death and ensuing vision loss. In this study, the role of p53 during retinal development and in the post-mitotic retina is investigated. A previously described super p53 transgenic mouse that expresses an extra copy of the mouse p53 gene driven by its endogenous promoter is utilized. Another transgenic mouse (HIP) that expresses the p53 gene in rod and cone photoreceptors driven by the human interphotoreceptor retinoid binding protein promoter was generated. The electroretinogram (ERG) of the super p53 mouse exhibited reduced rod-driven scotopic a and b wave and cone-driven photopic b wave responses. This deficit resulted from a reduced number of rod photoreceptors and inner nuclear layer cells. However, the reduced photopic signal arose only from lost inner retinal neurons, as cone numbers did not change. Furthermore, cell loss was non-progressive and resulted from increased apoptosis during retinal developmental as determined by TUNEL staining. In contrast, the continuous and specific expression of p53 in rod and cone photoreceptors in the mature retinas of HIP mice led to the selective loss of both rods and cones. These findings strongly support a role for p53 in regulating developmental apoptosis in the retina and suggest a potential role, either direct or indirect, for p53 in the degenerative photoreceptor loss associated with human blinding disorders.


Subject(s)
Apoptosis , Retinal Rod Photoreceptor Cells/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Shape , Cells, Cultured , Electroretinography , Evoked Potentials, Visual , Gene Expression , Humans , Mice , Mice, Transgenic , Retina/cytology , Retina/growth & development , Retina/metabolism
17.
BMC Neurosci ; 14: 70, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23844743

ABSTRACT

BACKGROUND: Phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac Exchanger 2 (P-Rex2) is a guanine nucleotide exchange factor (GEF) that specifically activates Rac GTPases, important regulators of actin cytoskeleton remodeling. P-Rex2 is known to modulate cerebellar Purkinje cell architecture and function, but P-Rex2 expression and function elsewhere in the central nervous system is unclear. To better understand potential roles for P-Rex2 in neuronal cytoskeletal remodeling and function, we performed widefield and confocal microscopy of specimens double immunolabeled for P-Rex2 and cell- and synapse-specific markers in the mouse retina. RESULTS: P-Rex2 was restricted to the plexiform layers of the retina and colocalized extensively with Vesicular Glutamate Transporter 1 (VGluT1), a specific marker for photoreceptor and bipolar cell terminals. Double labeling for P-Rex2 and peanut agglutinin, a cone terminal marker, confirmed that P-Rex2 was present in both rod and cone terminals. Double labeling with markers for specific bipolar cell types showed that P-Rex2 was present in the terminals of rod bipolar cells and multiple ON- and OFF-cone bipolar cell types. In contrast, P-Rex2 was not expressed in the processes or conventional synapses of amacrine or horizontal cells. CONCLUSIONS: P-Rex2 is associated specifically with the glutamatergic ribbon synaptic terminals of photoreceptors and bipolar cells that transmit visual signals vertically through the retina. The Rac-GEF function of P-Rex2 implies a specific role for P-Rex2 and Rac-GTPases in regulating the actin cytoskeleton in glutamatergic ribbon synaptic terminals of retinal photoreceptors and bipolar cells and appears to be ideally positioned to modulate the adaptive plasticity of these terminals.


Subject(s)
GTPase-Activating Proteins/metabolism , Neurons/cytology , Retina/cytology , Synapses/metabolism , Animals , Eye Proteins/metabolism , In Vitro Techniques , Lectins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/classification , Neurons/metabolism , Visual Pathways/cytology , Visual Pathways/metabolism
18.
Antioxid Redox Signal ; 19(16): 1877-88, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23682772

ABSTRACT

AIMS: Human γ-glutamyltranspeptidase 1 (hGGT1) is a cell-surface enzyme that is a regulator of redox adaptation and drug resistance due to its glutathionase activity. The human GGT2 gene encodes a protein that is 94% identical to the amino-acid sequence of hGGT1. Transcriptional profiling analyses in a series of recent publications have implicated the hGGT2 enzyme as a modulator of disease processes. However, hGGT2 has never been shown to encode a protein with enzymatic activity. The aim of this study was to express the protein encoded by hGGT2 and each of its known variants and to assess their stability, cellular localization, and enzymatic activity. RESULTS: We discovered that the proteins encoded by hGGT2 and its variants are inactive propeptides. We show that hGGT2 cDNAs are transcribed with a similar efficiency to hGGT1, and the expressed propeptides are N-glycosylated. However, they do not autocleave into heterodimers, fail to localize to the plasma membrane, and do not metabolize γ-glutamyl substrates. Substituting the coding sequence of hGGT1 to conform to alterations in a CX3C motif encoded by hGGT2 mRNAs disrupted autocleavage of the hGGT1 propeptide into a heterodimer, resulting in loss of plasma membrane localization and catalytic activity. INNOVATION AND CONCLUSIONS: This is the first study to evaluate hGGT2 protein. The data show that hGGT2 does not encode a functional enzyme. Microarray data which have reported induction of hGGT2 mRNA should not be interpreted as induction of a protein that has a role in the metabolism of extracellular glutathione and in maintaining the redox status of the cell.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Protein Processing, Post-Translational , Signal Transduction , gamma-Glutamyltransferase/metabolism , Amino Acid Sequence , Cell Membrane/enzymology , Cell Membrane/metabolism , Gene Expression Profiling/standards , HEK293 Cells , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/standards , Oxidation-Reduction , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Sequence Alignment , gamma-Glutamyltransferase/chemistry , gamma-Glutamyltransferase/genetics
19.
Int Rev Cell Mol Biol ; 300: 211-42, 2013.
Article in English | MEDLINE | ID: mdl-23273863

ABSTRACT

Melatonin is a chemical signal of darkness that is produced by retinal photoreceptors and pinealocytes. In the retina, melatonin diffuses from the photoreceptors to bind to specific receptors on a variety of inner retinal neurons to modify their activity. Potential target cells for melatonin in the inner retina are amacrine cells, bipolar cells, horizontal cells, and ganglion cells. Melatonin inhibits the release of dopamine from amacrine cells and increases the light sensitivity of horizontal cells. Melatonin receptor subtypes show differential, cell-specific patterns of expression that are likely to underlie differential functional modulation of specific retinal pathways. Melatonin potentiates rod signals to ON-type bipolar cells, via activation of the melatonin MT2 (Mel1b) receptor, suggesting that melatonin modulates the function of specific retinal circuits based on the differential distribution of its receptors. The selective and differential expression of melatonin receptor subtypes in cone circuits suggest a conserved function for melatonin in enhancing transmission from rods to second-order neurons and thus promote dark adaptation.


Subject(s)
Melatonin/physiology , Receptors, Melatonin/physiology , Retina/physiology , Animals , Circadian Rhythm , Gene Expression , Humans , Models, Biological , Neurotransmitter Agents/metabolism , Photoreceptor Cells, Vertebrate/physiology , Phylogeny , Pineal Gland/physiology , Receptors, Melatonin/genetics , Retina/cytology , Signal Transduction , Visual Pathways/cytology , Visual Pathways/physiology
20.
Cell Adh Migr ; 7(6): 469-75, 2013.
Article in English | MEDLINE | ID: mdl-24430202

ABSTRACT

Cell migration is fundamental to many biological processes, including development, normal tissue remodeling, wound healing, and many pathologies. However, cell migration is a complex process, and understanding its regulation in health and disease requires the ability to manipulate and measure this process quantitatively under controlled conditions. This report describes a simple in vitro assay for quantitative analysis of cell migration in two-dimensional cultures that is an inexpensive alternative to the classic "scratch" assay. The method described utilizes flexible silicone masks fabricated in the lab according to the research demands of the specific experiment to create a cell-free area for cells to invade, followed by quantitative analysis based on widely available microscopic imaging tools. This experimental approach has the important advantage of visualizing cell migration in the absence of the cellular damage and disruption of the substrate that occurs when the "wound" is created in the scratch assay. This approach allows the researcher to study the intrinsic migratory characteristics of cells in the absence of potentially confounding contributions from cellular responses to injury and disruption of cell-substrate interactions. This assay has been used with vascular smooth muscle cells, fibroblasts, and epithelial cell types, but should be applicable to the study of practically any type of cultured cell. Furthermore, this method can be easily adapted for use with fluorescence microscopy, molecular biological, or pharmacological manipulations to explore the molecular mechanisms of cell migration, live cell imaging, fluorescence microscopy, and correlative immunolabeling.


Subject(s)
Cell Movement/genetics , Fibroblasts/metabolism , Silicone Elastomers/chemistry , Biological Assay , Cells, Cultured , Humans , Silicone Elastomers/metabolism , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...