Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 324(6): E569-E576, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37166265

ABSTRACT

Classically, the regulation of energy balance has been based on central and peripheral mechanisms sensing energy, nutrients, metabolites, and hormonal cues. Several cellular mechanisms at central level, such as hypothalamic AMP-activated protein kinase (AMPK), integrate this information to elicit counterregulatory responses that control feeding, energy expenditure, and glucose homeostasis, among other processes. Recent data have added more complexity to the homeostatic regulation of metabolism by introducing, for example, the key role of "traditional" senses and sensorial information in this complicated network. In this regard, current evidence is showing that olfaction plays a key and bidirectional role in energy homeostasis. Although nutritional status dynamically and profoundly impacts olfactory sensitivity, the sense of smell is involved in food appreciation and selection, as well as in brown adipose tissue (BAT) thermogenesis and substrate utilization, with some newly described actors, such as olfactomedin 2 (OLFM2), likely playing a major role. Thus, olfactory inputs are contributing to the regulation of both sides of the energy balance equation, namely, feeding and energy expenditure (EE), as well as whole body metabolism. Here, we will review the current knowledge and advances about the role of olfaction in the regulation of energy homeostasis.


Subject(s)
Obesity , Smell , Humans , Obesity/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/metabolism , Energy Metabolism/physiology , Hypothalamus/metabolism , Thermogenesis
2.
Metabolism ; 129: 155122, 2022 04.
Article in English | MEDLINE | ID: mdl-35026233

ABSTRACT

BACKGROUND AND AIMS: Olfactomedin 2 (OLFM2; also known as noelin 2) is a pleiotropic protein that plays a major role in olfaction and Olfm2 null mice exhibit reduced olfactory sensitivity, as well as abnormal motor coordination and anxiety-related behavior. Here, we investigated the possible metabolic role of OLFM2. METHODS: Olfm2 null mice were metabolically phenotyped. Virogenetic modulation of central OLFM2 was also performed. RESULTS: Our data showed that, the global lack of OLFM2 in mice promoted anorexia and increased energy expenditure due to elevated brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT). This phenotype led to resistance to high fat diet (HFD)-induced obesity. Notably, virogenetic overexpression of Olfm2 in the lateral hypothalamic area (LHA) induced weight gain associated with decreased BAT thermogenesis. CONCLUSION: Overall, this evidence first identifies central OLFM2 as a new molecular actor in the regulation of whole-body energy homeostasis.


Subject(s)
Adipose Tissue, Brown , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Extracellular Matrix Proteins , Glycoproteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Thermogenesis/genetics
3.
Cells ; 10(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206213

ABSTRACT

The purpose of this study was to characterize the miRNA profile of purified retinal ganglion cells (RGC) from healthy and diseased rat retina. Diseased retina includes those after a traumatic optic nerve crush (ONC), and after ocular hypertension/glaucoma. Rats were separated into four groups: healthy/intact, 7 days after laser-induced ocular hypertension, 2 days after traumatic ONC, and 7 days after ONC. RGC were purified from rat retina using microbeads conjugated to CD90.1/Thy1. RNA were sequenced using Next Generation Sequencing. Over 100 miRNA were identified that were significantly different in diseased retina compared to healthy retina. Considerable differences were seen in the miRNA expression of RGC 7 days after ONC, whereas after 2 days, few changes were seen. The miRNA profiles of RGC 7 days after ONC and 7 days after ocular hypertension were similar, but discrete miRNA differences were still seen. Candidate mRNA showing different levels of expression after retinal injury were manipulated in RGC cultures using mimics/AntagomiRs. Of the five candidate miRNA identified and subsequently tested for therapeutic efficacy, miR-194 inhibitor and miR-664-2 inhibitor elicited significant RGC neuroprotection, whereas miR-181a mimic and miR-181d-5p mimic elicited significant RGC neuritogenesis.


Subject(s)
Glaucoma/genetics , Glaucoma/pathology , MicroRNAs/metabolism , Nerve Crush , Optic Nerve/pathology , Retinal Ganglion Cells/metabolism , Animals , Female , Gene Expression Regulation , Intraocular Pressure/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Neurites/metabolism , Neurogenesis/genetics , Neuroprotection/genetics , Ocular Hypertension/genetics , Ocular Hypertension/physiopathology , Rats, Sprague-Dawley
4.
Invest Ophthalmol Vis Sci ; 59(2): 995-1004, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29450543

ABSTRACT

Purpose: The compound heterozygous mutations in the ß subunit of chaperonin containing TCP-1 (CCT), encoded by CCT2, lead to the Leber congenital amaurosis (LCA). In this study, a cct2 mutant line of zebrafish was established to investigate the role of CCT2 mutations in LCA in vertebrates. Methods: A cct2 mutant zebrafish line was produced using the CRISPR-Cas9 system. Changes in the eyes of developing wild-type and mutant larvae were monitored using microscopy, immunostaining, TUNEL, and EdU assays. Phenotypic rescue of mutant phenotype was investigated by injection of CCT2 RNA into zebrafish embryos. Results: The cct2 mutation (L394H-7del) led to the synthesis of a mutated cctß protein with the L394H replacement and deletion of 7 amino acid residues (positions 395-401). The homozygous cct2-L394H-7del mutant exhibited a small eye phenotype at 2 days post fertilization (dpf) and was embryonically lethal after 5 dpf. In homozygous cct2-L394H-7del mutants, the retinal ganglion cell differentiation was attenuated, retinal cell cycle was affected, and the neural retinal cell death was significantly increased at 2 dpf compared with wild-type. Injection of RNA encoding wild-type human CCTß rescued the small eye phenotype, reduced retinal cell death, and restored the levels of CCTß protein and the major client protein Gß1 that were significantly reduced in the homozygous cct2-L394H-7del mutant compared with wild-type. These results indicate that cct2 plays an essential role in retinal development by regulating the cell cycle. Conclusions: The retinal pathology observed in the homozygous cct2-L394H-7del mutants resembles the retinal pathology of human LCA patients.


Subject(s)
Apoptosis/genetics , Cell Cycle/genetics , Chaperonin Containing TCP-1/genetics , Leber Congenital Amaurosis/genetics , Retina/pathology , Sequence Deletion , Zebrafish Proteins/genetics , Amino Acid Sequence , Animals , Blotting, Western , CRISPR-Cas Systems , Disease Models, Animal , Immunohistochemistry , In Situ Nick-End Labeling , Microscopy , Molecular Sequence Data , Phenotype , RNA/genetics , Zebrafish
5.
J Neurochem ; 143(6): 635-644, 2017 12.
Article in English | MEDLINE | ID: mdl-28975619

ABSTRACT

The olfm1a and olfm1b genes in zebrafish encode conserved secreted glycoproteins. These genes are preferentially expressed in the brain and retina starting from 16 h post-fertilization until adulthood. Functions of the Olfm1 gene is still unclear. Here, we produced and analyzed a null zebrafish mutant of both olfm1a and olfm1b genes (olfm1 null). olfm1 null fish were born at a normal Mendelian ratio and showed normal body shape and fertility as well as no visible defects from larval stages to adult. Olfm1 proteins were preferentially localized in the synaptosomes of the adult brain. Olfm1 co-immunoprecipitated with GluR2 and soluble NSF attachment protein receptor complexes indicating participation of Olfm1 in both pre- and post-synaptic events. Phosphorylation of GluR2 was not changed while palmitoylation of GluR2 was decreased in the brain synaptosomal membrane fraction of olfm1 null compared with wt fish. The levels of GluR2, SNAP25, flotillin1, and VAMP2 were markedly reduced in the synaptic microdomain of olfm1 null brain compared with wt. The internalization of GluR2 in retinal cells and the localization of VAMP2 in brain synaptosome were modified by olfm1 null mutation. This indicates that Olfm1 may regulate receptor trafficking from the intracellular compartments to the synaptic membrane microdomain, partly through the alteration of post-translational GluR2 modifications such as palmitoylation. Olfm1 may be considered a novel regulator of the composition and function of the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor complex.


Subject(s)
Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Protein Transport/genetics , Receptors, AMPA/metabolism , Animals , Brain/metabolism , Gene Knockout Techniques , Zebrafish
6.
Invest Ophthalmol Vis Sci ; 58(12): 5308-5318, 2017 10 01.
Article in English | MEDLINE | ID: mdl-29049729

ABSTRACT

Purpose: To elucidate functions of wild-type myocilin, a secreted glycoprotein associated with glaucoma. Methods: Lysates of mouse eyes were used for immunoprecipitation with affinity-purified antibodies against mouse myocilin. Shotgun proteomic analysis was used for the identification of proteins interacting with myocilin. Colocalization of myocilin and tissue inhibitor of metalloproteinases 3 (TIMP3) in different eye structures was investigated by a multiplex fluorescent in situ hybridization and immunofluorescent labeling with subsequent confocal microscopy. Matrix metalloproteinase 2 (MMP2) activity assay was used to test effects of myocilin on TIMP3 inhibitory action. Results: TIMP3 was identified by a shotgun proteomic analysis as a protein that was coimmunoprecipitated with myocilin from eye lysates of wild-type and transgenic mice expressing elevated levels of mouse myocilin but not from lysates of transgenic mice expressing mutated mouse myocilin. Interaction of myocilin and TIMP3 was confirmed by coimmunoprecipitation of myocilin and TIMP3 from HEK293 cells transiently transfected with cDNAs encoding these proteins. The olfactomedin domain of myocilin is essential for interaction with TIMP3. In the eye, the main sites of myocilin and TIMP3 colocalization are the trabecular meshwork, sclera, and choroid. Using purified proteins, it has been shown that myocilin markedly enhanced the inhibitory activity of TIMP3 toward MMP2. Conclusions: Myocilin may serve as a modulator of TIMP3 activity via interactions with the myocilin olfactomedin domain. Our data imply that in the case of MYOCILIN null or some glaucoma-causing mutations, inhibitory activity of TIMP3 toward MMP2 might be reduced, mimicking deleterious mutations in the TIMP3 gene.


Subject(s)
Cytoskeletal Proteins/physiology , Eye Proteins/physiology , Glycoproteins/physiology , Matrix Metalloproteinase 2/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Animals , Blotting, Western , Ciliary Body/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression , HEK293 Cells , Hep G2 Cells , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Proteomics , RNA, Messenger/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Trabecular Meshwork/metabolism , Transfection
7.
Invest Ophthalmol Vis Sci ; 58(11): 4703-4711, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28910446

ABSTRACT

Purpose: Our previous experiments demonstrated that intravitreal injection of platelet-derived growth factor-AA (PDGF-AA) provides retinal ganglion cell (RGC) neuroprotection in a rodent model of glaucoma. Here we used PDGFRα-enhanced green fluorescent protein (EGFP) mice to identify retinal cells that may be essential for RGC protection by PDGF-AA. Methods: PDGFRα-EGFP mice expressing nuclear-targeted EGFP under the control of the PDGFRα promoter were used. Localization of PDGFRα in the neural retina was investigated by confocal imaging of EGFP fluorescence and immunofluorescent labeling with a panel of antibodies recognizing different retinal cell types. Primary cultures of mouse RGCs were produced by immunopanning. Neurobiotin injection of amacrine cells in a flat-mounted retina was used for the identification of EGFP-positive amacrine cells in the inner nuclear layer. Results: In the mouse neural retina, PDGFRα was preferentially localized in the ganglion cell and inner nuclear layers. Immunostaining of the retina demonstrated that astrocytes in the ganglion cell layer and a subpopulation of amacrine cells in the inner nuclear layer express PDGFRα, whereas RGCs (in vivo or in vitro) did not. PDGFRα-positive amacrine cells are likely to be Type 45 gamma-aminobutyric acidergic (GABAergic) wide-field amacrine cells. Conclusions: These data indicate that the neuroprotective effect of PDGF-AA in a rodent model of glaucoma could be mediated by astrocytes and/or a subpopulation of amacrine cells. We suggest that after intravitreal injection of PDGF-AA, these cells secrete factors protecting RGCs.


Subject(s)
Amacrine Cells/metabolism , Astrocytes/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Retinal Ganglion Cells/drug effects , Animals , Biotin/analogs & derivatives , Biotin/pharmacology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Neuroprotection , Neuroprotective Agents , Retinal Ganglion Cells/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Arterioscler Thromb Vasc Biol ; 37(3): 446-454, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28062493

ABSTRACT

OBJECTIVE: The objective of this study is to investigate the role and underlying mechanism of Olfactomedin 2 (Olfm2) in smooth muscle cell (SMC) phenotypic modulation and vascular remodeling. APPROACH AND RESULTS: Platelet-derived growth factor-BB induces Olfm2 expression in primary SMCs while modulating SMC phenotype as shown by the downregulation of SMC marker proteins. Knockdown of Olfm2 blocks platelet-derived growth factor-BB-induced SMC phenotypic modulation, proliferation, and migration. Conversely, Olfm2 overexpression inhibits SMC marker expression. Mechanistically, Olfm2 promotes the interaction of serum response factor with the runt-related transcription factor 2 that is induced by platelet-derived growth factor-BB, leading to a decreased interaction between serum response factor and myocardin, causing a repression of SMC marker gene transcription and consequently SMC phenotypic modulation. Animal studies show that Olfm2 is upregulated in balloon-injured rat carotid arteries. Knockdown of Olfm2 effectively inhibits balloon injury-induced neointima formation. Importantly, knockout of Olfm2 in mice profoundly suppresses wire injury-induced neointimal hyperplasia while restoring SMC contractile protein expression, suggesting that Olfm2 plays a critical role in SMC phenotypic modulation in vivo. CONCLUSIONS: Olfm2 is a novel factor mediating SMC phenotypic modulation. Thus, Olfm2 may be a potential target for treating injury-induced proliferative vascular diseases.


Subject(s)
Carotid Artery Injuries/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Matrix Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Serum Response Factor/metabolism , Transcription Factors/metabolism , Vascular Remodeling , Animals , Aorta, Thoracic/metabolism , Becaplermin , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Nuclear Proteins/metabolism , Phenotype , Protein Binding , Proto-Oncogene Proteins c-sis/pharmacology , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , Trans-Activators/metabolism , Transfection , Vascular Remodeling/drug effects
9.
Nat Cell Biol ; 18(7): 765-76, 2016 07.
Article in English | MEDLINE | ID: mdl-27295555

ABSTRACT

To safeguard proteomic integrity, cells rely on the proteasome to degrade aberrant polypeptides, but it is unclear how cells remove defective proteins that have escaped degradation owing to proteasome insufficiency or dysfunction. Here we report a pathway termed misfolding-associated protein secretion, which uses the endoplasmic reticulum (ER)-associated deubiquitylase USP19 to preferentially export aberrant cytosolic proteins. Intriguingly, the catalytic domain of USP19 possesses an unprecedented chaperone activity, allowing recruitment of misfolded proteins to the ER surface for deubiquitylation. Deubiquitylated cargos are encapsulated into ER-associated late endosomes and secreted to the cell exterior. USP19-deficient cells cannot efficiently secrete unwanted proteins, and grow more slowly than wild-type cells following exposure to a proteasome inhibitor. Together, our findings delineate a protein quality control (PQC) pathway that, unlike degradation-based PQC mechanisms, promotes protein homeostasis by exporting misfolded proteins through an unconventional protein secretion process.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Protein Transport/physiology , Endopeptidases/deficiency , Endopeptidases/metabolism , Humans , Protein Binding , Proteomics/methods
10.
Hum Mol Genet ; 24(12): 3322-34, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25740847

ABSTRACT

Glaucoma is a multifactorial optic neuropathy characterized by retinal ganglion cell (RGC) death and axonal degeneration leading to irreversible blindness. Mutations in the myocilin (MYOC) gene are the most common genetic factors of primary open-angle glaucoma. To develop a genetic mouse model induced by the synergistic interaction of mutated myocilin and another significant risk factor, oxidative stress, we produced double-mutant mice (Tg-MYOC(Y437H/+)/Sod2(+/-)) bearing human MYOC with a Y437H point mutation and a heterozygous deletion of the gene for the primary antioxidant enzyme, superoxide dismutase 2 (SOD2). Sod2 is broadly expressed in most tissues including the trabecular meshwork (TM) and heterozygous Sod2 knockout mice exhibit the reduced SOD2 activity and oxidative stress in all studied tissues. Accumulation of Y437H myocilin in the TM induced endoplasmic reticulum stress and led to a 45% loss of smooth muscle alpha-actin positive cells in the eye drainage structure of 10- to 12-month-old Tg-MYOC(Y437H/+)/Sod2(+/-) mice as compared with wild-type littermates. Tg-MYOC(Y437H/+)/Sod2(+/-) mice had higher intraocular pressure, lost about 37% of RGCs in the peripheral retina, and exhibited axonal degeneration in the retina and optic nerve as compared with their wild-type littermates. Single-mutant littermates containing MYOC(Y437H/+) or Sod2(+/-) exhibited no significant pathological changes until 12 months of age. Additionally, we observed elevated expression of endothelial leukocyte adhesion molecule-1, a human glaucoma marker, in the TM of Tg-MYOC(Y437H/+)/Sod2(+/-) mice. This is the first reported animal glaucoma model that combines expression of a glaucoma-causing mutant gene and an additional mutation mimicking a deleterious environment factor that acts synergistically.


Subject(s)
Cytoskeletal Proteins/genetics , Epistasis, Genetic , Eye Proteins/genetics , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Heterozygote , Mutation , Superoxide Dismutase/deficiency , Actins/metabolism , Animals , Apoptosis/genetics , Axons/metabolism , Axons/pathology , Disease Models, Animal , E-Selectin/genetics , E-Selectin/metabolism , Endoplasmic Reticulum Stress/genetics , Gene Expression , Glaucoma, Open-Angle/diagnosis , Haploinsufficiency , Intraocular Pressure , Mice , Mice, Knockout , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Phenotype , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Trabecular Meshwork/metabolism
11.
Exp Eye Res ; 135: 127-33, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25720657

ABSTRACT

Adipose-derived mesenchymal stem cells (ASCs) hold promise for use in cell-based therapies. Their intrinsic anti-inflammatory properties are potentially useful for treatments of inflammatory conditions such as uveitis, while their ability to differentiate along multiple cell lineages suggests use in regenerating damaged or degenerated tissue. However, how ASCs will respond to the intraocular environment is poorly studied. We have recently reported that aqueous humor (AH), the fluid that nourishes the anterior segment of the eye, potently increases alkaline phosphatase (ALP) activity of ASCs, indicating osteogenic differentiation. Here, we expand on our previous findings to better define the nature of this response. To this end, we cultured ASCs in the presence of 0, 5, 10, and 20% AH and assayed them for ALP activity. We found ALP activity correlates with increasing AH concentrations from 5 to 20%, and that longer treatments result in increased ALP activity. By using serum free media and pretreating AH with dextran-coated charcoal, we found that serum and charcoal-adsorbable AH components augment but are not required for this response. Further, by heat-treating the AH, we established that thermally labile components are required for the osteogenic response. Finally, we showed myocilin, a protein present in AH, could induce ALP activity in ASCs. However, this was to a lesser extent than untreated 5% AH, and myocilin could only partially rescue the effect after heat treatment, documenting there were additional thermally labile constituents of AH involved in the osteogenic response. Our work adds to the understanding of the induction of ALP in ASCs following exposure to AH, providing important insight in how ASCs will be influenced by the ocular environment. In conclusion, increased osteogenic potential upon exposure to AH represents a potential challenge to developing ASC cell-based therapies directed at the eye.


Subject(s)
Adipose Tissue/cytology , Alkaline Phosphatase/metabolism , Aqueous Humor/physiology , Hot Temperature , Mesenchymal Stem Cells/enzymology , Osteogenesis/physiology , Analysis of Variance , Aqueous Humor/chemistry , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Cytoskeletal Proteins/pharmacology , Dose-Response Relationship, Drug , Eye Proteins/pharmacology , Glycoproteins/pharmacology , Humans
12.
Exp Neurol ; 261: 802-11, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25218043

ABSTRACT

Olfactomedin 2 (Olfm2) is a secretory glycoprotein belonging to the family of olfactomedin domain-containing proteins. A previous study has shown that a mutation in OLFM2 is associated with primary open angle glaucoma in Japanese patients. In the present study, we generated Olfm2 deficient mice by replacing the Olfm2 gene with the LacZ gene. The loss of Olfm2 resulted in no gross abnormalities. However, Olfm2 null mice showed reduced exploration, locomotion, olfactory sensitivity, abnormal motor coordination, and anxiety related behavior. The pattern of the Olfm2 gene expression was studied in the brain and eye using ß-galactosidase staining. In the brain, Olfm2 was mainly expressed in the olfactory bulb, cortex, piriform cortex, olfactory trabeculae, and inferior and superior colliculus. In the eye expression was detected mainly in retinal ganglion cells. In Olfm2 null mice, the amplitude of the first negative wave in the visual evoked potential test was significantly reduced as compared with wild-type littermates. Olfm2, similar to Olfm1, interacted with the GluR2 subunit of the AMPAR complexes and Olfm2 co-segregated with the AMPA receptor subunit GluR2 and other synaptic proteins in the synaptosomal membrane fraction upon biochemical fractionation of the adult mice cortex and retina. Immunoprecipitation from the synaptosomal membrane fraction of the Olfm2 null mouse brain cortex using the GluR2 antibody showed reduced levels of several components of the AMPAR complex in the immunoprecipitates including Olfm1, PSD95 and CNIH2. These results suggest that heterodimers of Olfm1 and Olfm2 interact with AMPAR more efficiently than Olfm2 homodimers and that Olfm2 plays a role in the organization of the AMPA receptor complexes.


Subject(s)
Extracellular Matrix Proteins/deficiency , Glycoproteins/deficiency , Movement Disorders/genetics , Olfaction Disorders/genetics , Receptors, AMPA/metabolism , Sequence Deletion/genetics , Vision Disorders/genetics , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Evoked Potentials, Visual/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Maze Learning/physiology , Mice , Mice, Knockout , Optic Nerve/pathology , Retina/metabolism , Vision Disorders/complications
13.
J Neurosci ; 34(16): 5539-51, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741044

ABSTRACT

Myocilin is a secreted glycoprotein that belongs to a family of olfactomedin domain-containing proteins. Although myocilin is detected in several ocular and nonocular tissues, the only reported human pathology related to mutations in the MYOCILIN gene is primary open-angle glaucoma. Functions of myocilin are poorly understood. Here we demonstrate that myocilin is a mediator of oligodendrocyte differentiation and is involved in the myelination of the optic nerve in mice. Myocilin is expressed and secreted by optic nerve astrocytes. Differentiation of optic nerve oligodendrocytes is delayed in Myocilin-null mice. Optic nerves of Myocilin-null mice contain reduced levels of several myelin-associated proteins including myelin basic protein, myelin proteolipid protein, and 2'3'-cyclic nucleotide 3'-phosphodiesterase compared with those of wild-type littermates. This leads to reduced myelin sheath thickness of optic nerve axons in Myocilin-null mice compared with wild-type littermates, and this difference is more pronounced at early postnatal stages compared with adult mice. Myocilin also affects differentiation of oligodendrocyte precursors in vitro. Its addition to primary cultures of differentiating oligodendrocyte precursors increases levels of tested markers of oligodendrocyte differentiation and stimulates elongation of oligodendrocyte processes. Myocilin stimulation of oligodendrocyte differentiation occurs through the NgR1/Lingo-1 receptor complex. Myocilin physically interacts with Lingo-1 and may be considered as a Lingo-1 ligand. Myocilin-induced elongation of oligodendrocyte processes may be mediated by activation of FYN and suppression of RhoA GTPase.


Subject(s)
Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Myelin Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/physiology , Optic Nerve/cytology , Receptors, Cell Surface/metabolism , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Glycoproteins/genetics , Humans , In Vitro Techniques , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation/genetics , Myelin Proteins/genetics , Nerve Tissue Proteins/genetics , Nogo Receptor 1 , Oligodendroglia/ultrastructure , Receptors, Cell Surface/genetics , Stem Cells/physiology
14.
J Biol Chem ; 289(14): 10155-67, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24563482

ABSTRACT

Myocilin, a causative gene for open angle glaucoma, encodes a secreted glycoprotein with poorly understood functions. To gain insight into its functions, we produced a stably transfected HEK293 cell line expressing myocilin under an inducible promoter and compared gene expression profiles between myocilin-expressing and vector control cell lines by a microarray analysis. A significant fraction of differentially expressed genes in myocilin-expressing cells was associated with cell growth and cell death, suggesting that myocilin may have a role in the regulation of cell growth and survival. Increased proliferation of myocilin-expressing cells was demonstrated by the WST-1 proliferation assay, direct cell counting, and immunostaining with antibodies against Ki-67, a cellular proliferation marker. Myocilin-containing conditioned medium also increased proliferation of unmodified HEK293 cells. Myocilin-expressing cells were more resistant to serum starvation-induced apoptosis than control cells. TUNEL-positive apoptotic cells were dramatically decreased, and two apoptotic marker proteins, cleaved caspase 7 and cleaved poly(ADP-ribose) polymerase, were significantly reduced in myocilin-expressing cells as compared with control cells under apoptotic conditions. In addition, myocilin-deficient mesenchymal stem cells exhibited reduced proliferation and enhanced susceptibility to serum starvation-induced apoptosis as compared with wild-type mesenchymal stem cells. Phosphorylation of ERK1/2 and its upstream kinases, c-Raf and MEK, was increased in myocilin-expressing cells compared with control cells. Elevated phosphorylation of ERK1/2 was also observed in the trabecular meshwork of transgenic mice expressing 6-fold higher levels of myocilin when compared with their wild-type littermates. These results suggest that myocilin promotes cell proliferation and resistance to apoptosis via the ERK1/2 MAPK signaling pathway.


Subject(s)
Cell Proliferation , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , MAP Kinase Signaling System/physiology , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/physiology , Caspase 7/genetics , Caspase 7/metabolism , Cell Survival/physiology , Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Glycoproteins/genetics , HEK293 Cells , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/physiology
16.
Brain ; 137(Pt 2): 503-19, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24176979

ABSTRACT

The development of neuroprotective strategies to attenuate retinal ganglion cell death could lead to novel therapies for chronic optic neuropathies such as glaucoma. Intravitreal transplantation of mesenchymal stem cells slows retinal ganglion cell death in models of optic nerve injury, but the mechanism of action remains unclear. Here we characterized the neuroprotective effects of mesenchymal stem cells and mesenchymal stem cell-derived factors in organotypic retinal explant culture and an in vivo model of ocular hypertensive glaucoma. Co-culture of rat and human bone marrow-derived mesenchymal stem cells with retinal explants increased retinal ganglion cell survival, after 7 days ex vivo, by ∼2-fold and was associated with reduced apoptosis and increased nerve fibre layer and inner plexiform layer thicknesses. These effects were not demonstrated by co-culture with human or mouse fibroblasts. Conditioned media from mesenchymal stem cells conferred neuroprotection, suggesting that the neuroprotection is mediated, at least partly, by secreted factors. We compared the concentrations of 29 factors in human mesenchymal stem cell and fibroblast conditioned media, and identified 11 enriched in the mesenchymal stem cell secretome. Treatment of retinal explants with a cocktail of these factors conferred retinal ganglion cell neuroprotection, with factors from the platelet-derived growth factor family being the most potent. Blockade of platelet-derived growth factor signalling with neutralizing antibody or with small molecule inhibitors of platelet-derived growth factor receptor kinase or downstream phosphatidylinositol 3 kinase eliminated retinal ganglion cell neuroprotection conferred by mesenchymal stem cell co-culture. Intravitreal injection of platelet-derived growth factor -AA or -AB led to profound optic nerve neuroprotection in vivo following experimental induction of elevated intraocular pressure. These data demonstrate that mesenchymal stem cells secrete a number of neuroprotective proteins and suggest that platelet-derived growth factor secretion in particular may play an important role in mesenchymal stem cell-mediated retinal ganglion cell neuroprotection. Furthermore, platelet-derived growth factor may represent an independent target for achieving retinal ganglion cell neuroprotection.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neuroprotective Agents/metabolism , Platelet-Derived Growth Factor/metabolism , Retinal Ganglion Cells/metabolism , Animals , Axotomy/adverse effects , Coculture Techniques/methods , Humans , Mesenchymal Stem Cells/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Ganglion Cells/pathology
17.
Exp Neurol ; 250: 205-18, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24095980

ABSTRACT

Olfactomedin 1 (Olfm1) is a secreted glycoprotein that is preferentially expressed in neuronal tissues. Here we show that deletion of exons 4 and 5 from the Olfm1 gene, which encodes a 52 amino acid long region in the N-terminal part of the protein, increased neonatal death and reduced body weight of surviving homozygous mice. Magnetic resonance imaging analyses revealed reduced brain volume and attenuated size of white matter tracts such as the anterior commissure, corpus callosum, and optic nerve. Adult Olfm1 mutant mice demonstrated abnormal behavior in several tests including reduced marble digging, elevated plus maze test, nesting activity and latency on balance beam tests as compared with their wild-type littermates. The olfactory system was both structurally and functionally disturbed by the mutation in the Olfm1 gene as shown by functional magnetic resonance imaging analysis and a smell test. Deficiencies of the olfactory system may contribute to the neonatal death and loss of body weight of Olfm1 mutant. Shotgun proteomics revealed 59 candidate proteins that co-precipitated with wild-type or mutant Olfm1 proteins in postnatal day 1 brain. Olfm1-binding targets included GluR2, Cav2.1, teneurin-4 and Kidins220. Modified interaction of Olfm1 with binding targets led to an increase in intracellular Ca(2+) concentration and activation of ERK1/2, MEK1 and CaMKII in the hippocampus and olfactory bulb of Olfm1 mutant mice compared with their wild-type littermates. Excessive activation of the CaMKII and Ras-ERK pathways in the Olfm1 mutant olfactory bulb and hippocampus by elevated intracellular calcium may contribute to the abnormal behavior and olfactory activity of Olfm1 mutant mice.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Brain/pathology , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Exons/genetics , Extracellular Matrix Proteins/genetics , Female , Glycoproteins/genetics , Immunoprecipitation , Magnetic Resonance Imaging , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
18.
J Biol Chem ; 288(37): 26357-71, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23897819

ABSTRACT

The glaucoma-associated gene, myocilin, is expressed in ocular and non-ocular tissues including the peripheral nervous system, but its functions in these tissues remain poorly understood. We demonstrate that in sciatic nerve, myocilin is expressed in Schwann cells with high concentrations at the nodes of Ranvier. There, myocilin interacts with gliomedin, neurofascin, and NrCAM, which are essential for node formation and function. Treatment of isolated dorsal root ganglion cultures with myocilin stimulates clustering of the nodal proteins neurofascin and sodium channel Nav1.2. Sciatic nerves of myocilin null mice express reduced levels of several myelin-associated and basal membrane proteins compared with those of wild-type littermates. They also demonstrate reduced myelin sheath thickness and partial disorganization of the nodes. Myocilin signaling through ErbB2/3 receptors may contribute to these observed effects. Myocilin binds to ErbB2/ErbB3, activates these receptors, and affects the downstream PI3K-AKT signaling pathway. These data implicate a role for myocilin in the development and/or maintenance of myelination and nodes of Ranvier in sciatic nerve.


Subject(s)
Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , Myelin Sheath/metabolism , Peripheral Nervous System/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Animals , Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Ganglia, Spinal/metabolism , Gene Expression Regulation , Glaucoma/metabolism , Glycoproteins/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Mutation , Myelin Sheath/genetics , Phosphorylation , Ranvier's Nodes/metabolism , Sciatic Nerve/metabolism , Signal Transduction
19.
J Biol Chem ; 288(23): 16882-16894, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23629661

ABSTRACT

Myocilin is a secreted glycoprotein that is expressed in ocular and non-ocular tissues. Mutations in the MYOCILIN gene may lead to juvenile- and adult-onset primary open-angle glaucoma. Here we report that myocilin is expressed in bone marrow-derived mesenchymal stem cells (MSCs) and plays a role in their differentiation into osteoblasts in vitro and in osteogenesis in vivo. Expression of myocilin was detected in MSCs derived from mouse, rat, and human bone marrow, with human MSCs exhibiting the highest level of myocilin expression. Expression of myocilin rose during the course of human MSC differentiation into osteoblasts but not into adipocytes, and treatment with exogenous myocilin further enhanced osteogenesis. MSCs derived from Myoc-null mice had a reduced ability to differentiate into the osteoblastic lineage, which was partially rescued by exogenous extracellular myocilin treatment. Myocilin also stimulated osteogenic differentiation of wild-type MSCs, which was associated with activation of the p38, Erk1/2, and JNK MAP kinase signaling pathways as well as up-regulated expression of the osteogenic transcription factors Runx2 and Dlx5. Finally, cortical bone thickness and trabecular volume, as well as the expression level of osteopontin, a known factor of bone remodeling and osteoblast differentiation, were reduced dramatically in the femurs of Myoc-null mice compared with wild-type mice. These data suggest that myocilin should be considered as a target for improving the bone regenerative potential of MSCs and may identify a new role for myocilin in bone formation and/or maintenance in vivo.


Subject(s)
Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , MAP Kinase Signaling System/physiology , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Cytoskeletal Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Eye Proteins/genetics , Glycoproteins/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Mutant Strains , Osteoblasts/cytology , Rats , Rats, Sprague-Dawley , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation/physiology
20.
J Biol Chem ; 287(44): 37171-84, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22923615

ABSTRACT

Olfm1, a secreted highly conserved glycoprotein, is detected in peripheral and central nervous tissues and participates in neural progenitor maintenance, cell death in brain, and optic nerve arborization. In this study, we identified Olfm1 as a molecule promoting axon growth through interaction with the Nogo A receptor (NgR1) complex. Olfm1 is coexpressed with NgR1 in dorsal root ganglia and retinal ganglion cells in embryonic and postnatal mice. Olfm1 specifically binds to NgR1, as judged by alkaline phosphatase assay and coimmunoprecipitation. The addition of Olfm1 inhibited the growth cone collapse of dorsal root ganglia neurons induced by myelin-associated inhibitors, indicating that Olfm1 attenuates the NgR1 receptor functions. Olfm1 caused the inhibition of NgR1 signaling by interfering with interaction between NgR1 and its coreceptors p75NTR or LINGO-1. In zebrafish, inhibition of optic nerve extension by olfm1 morpholino oligonucleotides was partially rescued by dominant negative ngr1 or lingo-1. These data introduce Olfm1 as a novel NgR1 ligand that may modulate the functions of the NgR1 complex in axonal growth.


Subject(s)
Axons/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Extracellular Matrix Proteins/physiology , Glycoproteins/physiology , Nerve Tissue Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Green Fluorescent Proteins/biosynthesis , Growth Cones/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins/physiology , Nogo Proteins , Optic Nerve/cytology , Optic Nerve/embryology , Organ Specificity , PC12 Cells , Protein Binding , Rats , Receptor, Nerve Growth Factor/metabolism , Zebrafish , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...