Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 289(8): 4660-73, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24371137

ABSTRACT

The subcellular distribution of the G protein-coupled receptor GPR37 affects cell viability and is implicated in the pathogenesis of parkinsonism. Intracellular accumulation and aggregation of GPR37 cause cell death, whereas GPR37 located in the plasma membrane provides cell protection. We define here a pathway through which the recently identified natural ligand, prosaposin, promotes plasma membrane association of GPR37. Immunoabsorption of extracellular prosaposin reduced GPR37(tGFP) surface density and decreased cell viability in catecholaminergic N2a cells. We found that GPR37(tGFP) partitioned in GM1 ganglioside-containing lipid rafts in the plasma membrane of live cells. This partitioning required extracellular prosaposin and was disrupted by lipid raft perturbation using methyl-ß-cyclodextrin or cholesterol oxidase. Moreover, complex formation between GPR37(tGFP) and the GM1 marker cholera toxin was observed in the plasma membrane. These data show functional association between GPR37, prosaposin, and GM1 in the plasma membrane. These results thus tie together the three previously defined components of the cellular response to insult. Our findings identify a mechanism through which the receptor's natural ligand and GM1 may protect against toxic intracellular GPR37 aggregates observed in parkinsonism.


Subject(s)
Cell Membrane/metabolism , G(M1) Ganglioside/metabolism , Receptors, G-Protein-Coupled/metabolism , Saposins/metabolism , Animals , Cell Membrane/drug effects , Cell Polarity/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Extracellular Space/metabolism , Flow Cytometry , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Microdomains/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Growth Factors/metabolism , Peptides/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Time Factors , beta-Cyclodextrins/pharmacology
2.
Cent Nerv Syst Agents Med Chem ; 9(2): 119-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-20021345

ABSTRACT

Prosaposin, a 66 kDa glycoprotein, was identified initially as the precursor of the sphingolipid activator proteins, saposins A-D, which are required for the enzymatic hydrolysis of certain sphingolipids by lysosomal hydrolases. While mature saposins are distributed to lysosomes, prosaposin exists in secretory body fluids and plasma membranes. In addition to its role as the precursor, prosaposin shows a variety of neurotrophic and myelinotrophic activities through a receptor-mediated mechanism. In studies in vivo, prosaposin was demonstrated to exert a variety of neuro-efficacies capable of preventing neuro-degeneration following neuro-injury and promoting the amelioration of allodynia and hyperalgesia in pain models. Collective findings indicate that prosaposin is not a simple house-keeping precursor protein; instead, it is a protein essentially required for the development and maintenance of the central and peripheral nervous systems. Accumulating evidence over the last decade has attracted interests in exploring and developing new therapeutic approaches using prosaposin for human disorders associated with neuro-degeneration. In this review we detail the structure characteristics, cell biological feature, in vivo efficacy, and neuro-therapeutic potential of prosaposin, thereby providing future prospective in clinical application of this multifunctional protein.


Subject(s)
Biochemistry , Biological Transport/physiology , Glycoproteins/therapeutic use , Neurobiology , Peripheral Nervous System/physiology , Protein Precursors/metabolism , Saposins/physiology , Sphingolipid Activator Proteins/therapeutic use , Alternative Splicing/genetics , Amino Acid Sequence , Conserved Sequence/genetics , Conserved Sequence/physiology , Female , Humans , Lactation/metabolism , Lipid Metabolism/physiology , Molecular Sequence Data , Nerve Growth Factors/physiology , Protein Precursors/genetics , Saposins/genetics , Saposins/metabolism , Sphingolipid Activator Proteins/metabolism , Tissue Distribution/physiology , Transfection
3.
Proc Natl Acad Sci U S A ; 106(42): 17717-22, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815502

ABSTRACT

The pleiotropic signaling lipid sphingosine-1-phosphate (S1P) plays significant roles in angiogenesis, heart disease, and cancer. LT1009 (also known as sonepcizumab) is a humanized monoclonal antibody that binds S1P with high affinity and specificity. Because the antibody is currently in clinical trials, it is important to confirm by structural and biochemical analyses that it binds its target in a predictable manner. Therefore, we determined the structure of a complex between the LT1009 antibody Fab fragment and S1P refined to 1.90 A resolution. The antibody employs unique and diverse strategies to recognize its antigen. Two metal ions bridge complementarity determining regions from the antibody light chain and S1P. The coordination geometry, inductively coupled plasma spectroscopy, surface plasmon resonance spectroscopy, and biochemical assays suggest that these are Ca(2+). The amino alcohol head group of the sphingosine backbone is recognized through hydrogen bonding interactions from 1 aa side chain and polypeptide backbone atoms of the antibody light and heavy chains. The S1P hydrophobic tail is almost completely enclosed within a hydrophobic channel formed primarily by the heavy chain. Both treatment of the complex with metal chelators and mutation of amino acids in the light chain that coordinate the metal atoms or directly contact the polar head group abrogate binding, while mutations within the hydrophobic cavity also decrease S1P binding affinity. The structure suggests mechanistic details for recognition of a signaling lipid by a therapeutic antibody candidate. Moreover, this study provides direct structural evidence that antibodies are capable of using metals to bridge antigen:antibody complexes.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigen-Antibody Complex/chemistry , Immunoglobulin Fab Fragments/chemistry , Lysophospholipids/chemistry , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , Animals , Antibodies, Monoclonal/genetics , Antibody Affinity , Antibody Specificity , Binding Sites, Antibody/genetics , Calcium/chemistry , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/genetics , In Vitro Techniques , Lysophospholipids/antagonists & inhibitors , Mice , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Sphingosine/antagonists & inhibitors , Sphingosine/chemistry , Sphingosine/immunology , Surface Plasmon Resonance
4.
J Lipid Res ; 50(11): 2245-57, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19509417

ABSTRACT

Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid involved in multiple physiological processes. Importantly, dysregulated S1P levels are associated with several pathologies, including cardiovascular and inflammatory diseases and cancer. This report describes the successful production and characterization of a murine monoclonal antibody, LT1002, directed against S1P, using novel immunization and screening methods applied to bioactive lipids. We also report the successful generation of LT1009, the humanized variant of LT1002, for potential clinical use. Both LT1002 and LT1009 have high affinity and specificity for S1P and do not cross-react with structurally related lipids. Using an in vitro bioassay, LT1002 and LT1009 were effective in blocking S1P-mediated release of the pro-angiogenic and prometastatic cytokine, interleukin-8, from human ovarian carcinoma cells, showing that both antibodies can out-compete S1P receptors in binding to S1P. In vivo anti-angiogenic activity of all antibody variants was demonstrated using the murine choroidal neovascularization model. Importantly, intravenous administration of the antibodies showed a marked effect on lymphocyte trafficking. The resulting lead candidate, LT1009, has been formulated for Phase 1 clinical trials in cancer and age-related macular degeneration. The anti-S1P antibody shows promise as a novel, first-in-class therapeutic acting as a "molecular sponge" to selectively deplete S1P from blood and other compartments where pathological S1P levels have been implicated in disease progression or in disorders where immune modulation may be beneficial.


Subject(s)
Antibodies, Monoclonal/immunology , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/pharmacology , Antibody Specificity/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Choroidal Neovascularization/prevention & control , Cross Reactions/immunology , Female , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/metabolism , Interleukins/metabolism , Kinetics , Lysophospholipids/metabolism , Macular Degeneration/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Mutagenesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding , Signal Transduction/drug effects , Signal Transduction/immunology , Sphingosine/immunology , Sphingosine/metabolism , Surface Plasmon Resonance
5.
FEBS J ; 275(19): 4903-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18761669

ABSTRACT

Prosaposin is a neurotrophic factor that has been demonstrated to mediate trophic signalling events in different cell types; it distributes to surface membranes of neural cells and also exists as a secreted protein in different body fluids. Prosaposin was demonstrated to form tightly bound complexes with a variety of gangliosides, and a functional role has been suggested for ganglioside-prosaposin complexes. In this work, we provide evidence that exogenous prosaposin triggers a signal cascade after binding to its target molecules on lipid rafts of pheochromocytoma PC12 cell plasma membranes, as revealed by scanning confocal microscopy and linear sucrose gradient analysis. In these cells, prosaposin is able to induce extracellular signal-regulated kinase phosphorylation, sphingosine kinase activation, and consequent cell death prevention, acting through lipid rafts. These findings point to the role of lipid rafts in the prosaposin-triggered signalling pathway, thus supporting a role for this factor as a new component of the multimolecular signalling complex involved in the neurotrophic response.


Subject(s)
Membrane Microdomains/physiology , Saposins/physiology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , PC12 Cells , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats
6.
Biochim Biophys Acta ; 1780(12): 1441-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18706485

ABSTRACT

Prosaposin triggers G-protein-coupled receptor (GPCR)-mediated protein kinase B (Akt)/extracellular signal-regulated kinase (ERK) phosphorylation cascades to exert its neurotrophic and myelinotrophic activity capable of preventing neural cell death and promoting neural proliferation and glial differentiation. In the present study, we investigated the down-stream neurotrophic signaling mechanism of prosaposin by which rat pheochromocytoma (PC-12) cells are protected from cell death induced by oxidative stress. When PC-12 cells were exposed to H2O2, the cells underwent abrupt shrinkage followed by apoptosis. Prosaposin treatment at as low as 1 nM protected PC-12 cells from cell death by the oxidative stress with the activation of an ERK phosphorylation cascade. Simultaneously, prosaposin blocked the oxidative stress induced-Akt phosphorylation that acts on the down-stream of caspase-3 activation. A MEK inhibitor, PD98059, or a phosphatidylinositol 3-kinase (PI3K) inhibitor, LY294002, abolished the survival effect of prosaposin on the oxidative stress-induced cell death. Furthermore, prosaposin blocked the oxidative stress-induced phosphorylations of c-Jun N-terminal kinase (JNK) and p38 stress-activated protein kinase. We further investigated the effect of prosaposin treatment on the phosphorylation of activating protein-1 (AP-1) complex components, c-Jun and activating transcription factor (ATF)-3. Western blot analysis demonstrated that prosaposin treatment at 100 ng/ml decreased the levels of c-Jun and ATF-3 induced by H2O2 stimulation. Our results suggest that prosaposin aids survival of PC-12 cells from oxidative stress not only by reducing the phosphorylation levels of JNK and p38, but also by regulating the c-Jun/AP-1 pathway.


Subject(s)
Activating Transcription Factor 3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/physiology , Oxidative Stress , Saposins/physiology , Animals , Caspase 3/physiology , Cell Death/physiology , Cell Survival/physiology , Dimerization , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Genes Chromosomes Cancer ; 44(4): 351-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16080200

ABSTRACT

We identified prosaposin (PSAP) as a secreted protein expressed in androgen-independent (AI) prostate cancer cells by cloning/sequencing, after probing a PC-3 cDNA library expressed in the lambdaTriplEx phagemid expression vector with a polyclonal rabbit antibody generated against pooled human seminal plasma. PSAP is a neurotrophic molecule; its deficiency or inactivation has proved to be lethal in man and mice, and in mice, it leads to abnormal development and atrophy of the prostate gland, despite normal testosterone levels. We used Southern hybridization, quantitative real-time polymerase chain reaction, and/or single nucleotide polymorphism (SNP) array analysis, and we now report the genomic amplification of PSAP in the metastatic AI prostate cancer cell lines, PC-3, DU-145, MDA-PCa 2b, M-12, and NCI-H660. In addition, by using SNP arrays and a set of 25 punch biopsy samples of human prostate cancer xenografts (LAPC3, LuCaP 23.1, 35, 49, 58, 73, 77, 81, 86.2, 92.1, 93, 96, 105, and 115), lymph nodes, and visceral-organ metastases, we detected amplification of the PSAP locus (10q22.1) in LuCaP 58 and 96 xenografts and two lymph node metastases. In addition, AI metastatic prostate cancer cell lines C4-2B and IA8-ARCaP over-expressed PSAP mRNA without evidence of genomic amplification. Taken together with prior data that demonstrated the growth-, migration-, and invasion-promoting activities, the activation of multiple signal transduction pathways, and the antiapoptotic effect of PSAP (or one of its active domains, saposin C) in prostate cancer cells, our current observation of PSAP amplification or overexpression in prostate cancer suggests, for the first time, a role for this molecule in the process of carcinogenesis or cancer progression in the prostate.


Subject(s)
Gene Amplification , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Saposins/genetics , Saposins/metabolism , Blotting, Western , Cell Line, Tumor , Cloning, Molecular , Genes, Neoplasm , Humans , In Situ Hybridization , Male , Microarray Analysis , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Saposins/chemistry
8.
Asian J Androl ; 7(2): 147-58, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15897971

ABSTRACT

AIM: To determine the effect of saposin C (a known trophic domain of prosaposin) on proliferation, migration and invasion, as well as its effect on the expression of urokinase plasmonogen activator (uPA), its receptor (uPAR) and matrix metalloproteinases (MMP)-2 and -9 in normal and malignant prostate cells. In addition, we tested whether saposin C can activate p42/44 and stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) signal transduction pathways of the mitogen-activated protein kinase (MAPK) superfamily. METHODS: We employed Western blot analysis, phospho-specific antibodies, cell proliferation assay, reverse transcriptase-polymerase chain reaction, in vitro kinase assays and migration and invasion to determine the effect of saposin C on various biological behaviors of prostate stromal and cancer cells. RESULTS: Saposin C, in a cell type-specific manner, upregulates uPA/uPAR and immediate early gene c-Jun expression, stimulates cell proliferation, migration and invasion and activates p42/44 and SAPK/JNK MAPK pathways in prostate stromal and cancer cells. Normal prostate epithelial cells were not responsive to saposin C treatment in the above studies. CONCLUSION: Saposin C functions as a multipotential modulator of diverse biological activities in prostate cancer and stromal cells. These results strongly suggest that saposin C functions as a potent growth factor for prostatic cells and may contribute to prostate carcinogenesis and/or the development of hormone-refractory prostate cancer.


Subject(s)
Cell Division/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Receptors, Cell Surface/genetics , Saposins/pharmacology , Signal Transduction , Stromal Cells/metabolism , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics , Enzyme Activation , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Urokinase Plasminogen Activator , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/enzymology , Stromal Cells/pathology
9.
Prostate ; 61(2): 114-23, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15305334

ABSTRACT

BACKGROUND: Prosaposin is a neurotrophic factor. Prosaposin knock-out mice have been reported to develop a number of abnormalities, including atrophy of the prostate gland and mitogen-activated protein kinase (MAPK)-inactivation in prostate epithelial cells. These abnormalities underscore a potential fundamental role in prostate development. The trophic factor activity of prosaposin has been localized at a specific amino terminal portion of the molecule that has been the source for a number of biologically active peptides called prosaptides (e.g., TX14A). The expression and function of prosaposin in prostate cancer is not known. METHODS: Using conventional protein expression analysis, immunohistochemical staining, cell proliferation assays, and in vitro invasion assays, we determined the expression of prosaposin and the effect of prosaptide TX14A on cell growth/death protection, motility, invasion, and MAPK signal transduction pathway in prostate cancer cells. RESULTS: We found a higher expression of prosaposin in androgen-independent (AI) prostate cancer cells (PC-3 and DU-145) than in androgen-dependent (AD) LNCaP or normal prostate epithelial cells. Immunohistochemical staining on benign and malignant prostate tissues revealed an intense cytoplasmic anti-prosaposin immunoreactivity in tumor cells, as well as stromal, endothelial, and inflammatory mononuclear cells. The intensity of staining was proportional to the overall Gleason's score. In addition, we demonstrated that TX14A stimulates cell proliferation/survival, migration, and invasion, and activates the Raf-MEK-ERK-RSK-Elk-1 signaling cascade of the MAPK pathway. CONCLUSIONS: These results are suggestive of a potential pleuripotent regulatory function for prosaposin in prostate cancer.


Subject(s)
Cell Movement/drug effects , Glycoproteins/metabolism , MAP Kinase Signaling System/drug effects , Nerve Growth Factors/pharmacology , Prostatic Neoplasms/physiopathology , Cell Division/drug effects , Cell Line, Tumor , Humans , Male , Neoplasm Invasiveness/physiopathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Saposins
10.
Exp Cell Res ; 298(1): 38-47, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15242760

ABSTRACT

We report that prosaposin binds to U937 and is active as a protective factor on tumor necrosis factor alpha (TNFalpha)-induced cell death. The prosaposin-derived saposin C binds to U937 cells in a concentration-dependent manner, suggesting that prosaposin behaves similarly. Prosaposin binding induces U937 cell death prevention, reducing both necrosis and apoptosis. This effect was inhibited by mitogen-activated protein ERK kinase (MEK) and sphingosine kinase (SK) inhibitors, indicating that prosaposin prevents cell apoptosis by activation of extracellular signal-regulated kinases (ERKs) and sphingosine kinase. Prosaposin led to rapid ERK phosphorylation in U937 cells as detected by anti-phospho-p44/42 mitogen-activated protein (MAP) kinase and anti-phosphotyrosine reactivity on ERK immunoprecipitates. It was partially prevented by apo B-100 and pertussis toxin (PT), suggesting that both lipoprotein receptor-related protein (LRP) receptor and Go-coupled receptor may play a role in the prosaposin-triggered pathway. Moreover, sphingosine kinase activity was increased by prosaposin treatment as demonstrated by the enhanced intracellular formation of sphingosine-1-phosphate (S-1-P). The observation that the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin prevented the prosaposin effect on cell apoptosis suggests that sphingosine kinase exerts its anti-apoptotic activity by the PI3K-Akt pathway. Thus, cell apoptosis prevention by prosaposin occurs through ERK phosphorylation and sphingosine kinase. The biological effect triggered by prosaposin might be extended to primary cells because it triggers Erk phosphorylation in peripheral blood mononuclear cells (PBMCs). This is the first evidence of a biological effect consequent to a signal transduction pathway triggered by prosaposin in cells of non-neurological origin.


Subject(s)
Apoptosis/immunology , Glycoproteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Cell Survival/immunology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Glycoproteins/pharmacology , Humans , Lysophospholipids/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monocytes/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Binding/physiology , Receptors, LDL/metabolism , Saposins , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
11.
Hippocampus ; 14(3): 275-80, 2004.
Article in English | MEDLINE | ID: mdl-15132426

ABSTRACT

Several studies indicate that stress can produce remarkable effects on neurotrophic factors. In this regard, hippocampus is the most interesting structure of the brain because of its broad involvement in behavioral and neuroendocrine phenomena. In the present study, we investigated the effect of stress on hippocampal prosaposin, which is known to act as a neurotrophic and neuroprotective factor. Rats subjected to restraint stress (120 min) had a significant and transient reduction of hippocampal, but not hypothalamic, prosaposin full-length protein. Indeed, when this stressful stimulus was applied daily for 3 days, no differences were detected in comparison with naive rats. To investigate the role of glucocorticoids in the stress-induced decrease in hippocampal prosaposin, adrenalectomized and corticosterone-treated rats were studied. The results indicate that adrenalectomized rats behave as intact animals. This finding indicates that the absence of endogenous corticosterone does not prevent a decrease in hippocampal prosaposin. When an increase of corticosterone was achieved through exogenous administration, hippocampal prosaposin concentrations were unchanged in comparison with vehicle-injected (sesame oil) rats. These results led to the conclusion that stress, not via an increase of glucocorticoid hormone, transiently reduces hippocampal prosaposin levels. This phenomenon is followed by rapid recovery of the neurotrophin level, even when the stress stimulus persists.


Subject(s)
Down-Regulation/physiology , Glucocorticoids/physiology , Glycoproteins/metabolism , Hippocampus/metabolism , Nerve Growth Factors/pharmacology , Stress, Physiological/metabolism , Adrenalectomy , Animals , Corticosterone/pharmacology , Corticosterone/physiology , Down-Regulation/drug effects , Glucocorticoids/pharmacology , Hippocampus/drug effects , Hippocampus/physiopathology , Hypothalamus/metabolism , Male , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology , Saposins , Stress, Physiological/physiopathology
12.
J Neurotrauma ; 20(8): 755-65, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12965054

ABSTRACT

Prosaposin, the precursor of saposins or saps, is an injury-repair protein that acts on both neurons and glia. Previous studies identified the prosaposin gene as one of differentially expressed genes following nerve injury. In the present study, we investigated expression of prosaposin mRNA in injured brain utilizing rat models of focal cerebral ischemia and cortical stab wound in order to explore the significance of prosaposin in nerve injury. In ischemic brain, the level of prosaposin mRNA was elevated greater than 400% over controls within 5 days after ischemic insults. Importantly, this induction was accompanied by a 9-base splicing consistent with the alternative Exon-8 splicing of human prosaposin mRNA. In normal brain, two prosaposin mRNA species with and without the 9-base insertion were expressed at a ratio of 85:15; however, this equilibrium reverted to 5:95 following ischemic injury. Similar inductions were observed in stab wound brains. Immunohistochemical staining and in situ hybridization demonstrated an enhanced signal distribution of prosaposin mRNA and injury-induced prosaposin protein around the lesion. The data suggest the expression and processing of prosaposin mRNA may be crucially regulated not only for cerebral homeostasis but also during nerve regenerative and degenerative processes.


Subject(s)
Brain Injuries/genetics , Gene Expression Regulation , Glycoproteins/biosynthesis , RNA, Messenger/genetics , Alternative Splicing/genetics , Animals , Blotting, Northern , Brain Injuries/metabolism , Exons/genetics , Gene Library , Immunohistochemistry , In Situ Hybridization , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Saposins
SELECTION OF CITATIONS
SEARCH DETAIL
...