Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 373: 114656, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38114054

ABSTRACT

Studies have reported that Prosaposin (PSAP) is neuroprotective in cerebrovascular diseases. We hypothesized that PSAP would reduce infarct volume by attenuating neuronal apoptosis and promoting cell survival through G protein-coupled receptor 37(GPR37)/PI3K/Akt/ASK1 pathway in middle cerebral artery occlusion (MCAO) rats. Two hundred and thirty-five male and eighteen female Sprague-Dawley rats were used. Recombinant human PSAP (rPSAP) was administered intranasally 1 h (h) after reperfusion. PSAP small interfering ribonucleic acid (siRNA), GPR37 siRNA, and PI3K specific inhibitor LY294002 were administered intracerebroventricularly 48 h before MCAO. Infarct volume, neurological score, immunofluorescence staining, Western blot, Fluoro-Jade C (FJC) and TUNEL staining were examined. The expression of endogenous PSAP and GPR37 were increased after MCAO. Intranasal administration of rPSAP reduced brain infarction, neuronal apoptosis, and improved both short- and long-term neurological function. Knockdown of endogenous PSAP aggravated neurological deficits. Treatment with exogenous rPSAP increased PI3K expression, Akt and ASK1 phosphorylation, and Bcl-2 expression; phosphorylated-JNK and Bax levels were reduced along with the number of FJC and TUNEL positive neurons. GPR37 siRNA and LY294002 abolished the anti-apoptotic effect of rPSAP at 24 h after MCAO. In conclusion, rPSAP attenuated neuronal apoptosis and improved neurological function through GPR37/PI3K/Akt/ASK1 pathway after MCAO in rats. Therefore, further exploration of PSAP as a potential treatment option in ischemic stroke is warranted.


Subject(s)
Neuroprotective Agents , Proto-Oncogene Proteins c-akt , Rats , Male , Female , Humans , Animals , Rats, Sprague-Dawley , Proto-Oncogene Proteins c-akt/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Saposins/metabolism , Saposins/pharmacology , Saposins/therapeutic use , Signal Transduction , Administration, Intranasal , Apoptosis , RNA, Small Interfering/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
2.
Anat Sci Int ; 96(3): 359-369, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33534127

ABSTRACT

Prosaposin (PS) is the precursor of four sphingolipid activator proteins, saposin A-D. PS is both a precursor protein and a neuroprotective factor, and is up-regulated in response to excitotoxicity induced by kainic acid (KA), a glutamate analogue. Excess glutamate release induces neuropathological disorders such as ischemia and seizure. Our group's research revealed that PS immunoreactivity (IR) increased significantly in the hippocampal and cortical neurons on day 3 after KA injection, and high PS levels were maintained even after 3 weeks. The increase in PS, but not saposins, as detected by immunoblotting, suggests that the increase in PS-IR after KA injection was not caused by an increase in saposins acting as lysosomal enzymes after neuronal damage but, rather, by an increase in PS as a neurotrophic factor to improve neuronal survival. An 18-mer peptide (PS18) derived from the PS neurotrophic region significantly protected hippocampal neurons against KA-induced destruction. Furthermore, parvalbumin-positive GABAergic inhibitory interneurons and their axons exhibited intense PS expression. These results suggest that axonally transported PS protects damaged hippocampal pyramidal neurons from KA-induced neurotoxicity. Further in vitro studies that include the transfection of the PS gene will help with clarifying the mechanisms underlying the transport and secretion of PS.


Subject(s)
Kainic Acid/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Saposins/pharmacology , Animals , Rats
3.
Article in English | MEDLINE | ID: mdl-29662839

ABSTRACT

Due to their archaic life style and microbivor behavior, amoebae may represent a source of antimicrobial peptides and proteins. The amoebic protozoon Dictyostelium discoideum has been a model organism in cell biology for decades and has recently also been used for research on host-pathogen interactions and the evolution of innate immunity. In the genome of D. discoideum, genes can be identified that potentially allow the synthesis of a variety of antimicrobial proteins. However, at the protein level only very few antimicrobial proteins have been characterized that may interact directly with bacteria and help in fighting infection of D. discoideum with potential pathogens. Here, we focus on a large group of gene products that structurally belong to the saposin-like protein (SAPLIP) family and which members we named provisionally Apls (amoebapore-like peptides) according to their similarity to a comprehensively studied antimicrobial and cytotoxic pore-forming protein of the protozoan parasite Entamoeba histolytica. We focused on AplD because it is the only Apl gene that is reported to be primarily transcribed further during the multicellular stages such as the mobile slug stage. Upon knock-out (KO) of the gene, aplD- slugs became highly vulnerable to virulent Klebsiella pneumoniae. AplD- slugs harbored bacterial clumps in their interior and were unable to slough off the pathogen in their slime sheath. Re-expression of AplD in aplD- slugs rescued the susceptibility toward K. pneumoniae. The purified recombinant protein rAplD formed pores in liposomes and was also capable of permeabilizing the membrane of live Bacillus megaterium. We propose that the multifarious Apl family of D. discoideum comprises antimicrobial effector polypeptides that are instrumental to interact with bacteria and their phospholipid membranes. The variety of its members would allow a complementary and synergistic action against a variety of microbes, which the amoeba encounters in its environment.


Subject(s)
Bacterial Infections/immunology , Dictyostelium/immunology , Dictyostelium/microbiology , Host-Pathogen Interactions/immunology , Immunity, Innate , Saposins/metabolism , Saposins/pharmacology , Animals , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Bacillus megaterium/drug effects , Dictyostelium/genetics , Dictyostelium/metabolism , Gastropoda/immunology , Gastropoda/metabolism , Gastropoda/microbiology , Gene Expression Profiling , Ion Channels/metabolism , Ion Channels/pharmacology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/pathogenicity , Liposomes/metabolism , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Protozoan Proteins/metabolism , Protozoan Proteins/pharmacology , Recombinant Proteins , Saposins/genetics , Saposins/immunology
4.
Curr Protein Pept Sci ; 18(7): 758-764, 2017.
Article in English | MEDLINE | ID: mdl-26965692

ABSTRACT

Gaucher disease is an autosomal recessive lysosomal storage disorder, caused by mutations in the GBA gene. The frequency of Gaucher disease patients and heterozygote carriers that developed Parkinson disease has been found to be above that of the control population. This fact suggests that mutations in the GBA gene can be involved in Parkison's etiology. Analysis of large cohorts of patients with Parkinson disease has shown that there are significantly more cases bearing GBA mutations than those found among healthy individuals. Functional studies have proven an interaction between α-synuclein and GBA, the levels of which presented an inverse correlation. Mutant GBA proteins cause increases in α-synuclein levels, while an inhibition of GBA by α-synuclein has been also demonstrated. Saposin C, a coactivator of GBA, has been shown to protect GBA from this inhibition. Among the GBA variants associated with Parkinson disease, E326K seems to be one of the most prevalent. Interestingly, it is involved in Gaucher disease only when it forms part of a double-mutant allele, usually with the L444P mutation. Structural analyses have revealed that both residues (E326 and L444) interact with Saposin C and, probably, also with α-synuclein. This could explain the antagonistic role of these two proteins in relation to GBA.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Mutation , Parkinson Disease/genetics , alpha-Synuclein/genetics , Alleles , Gaucher Disease/complications , Gaucher Disease/metabolism , Gaucher Disease/pathology , Gene Expression , Gene Frequency , Glucosylceramidase/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Parkinson Disease/complications , Parkinson Disease/metabolism , Parkinson Disease/pathology , Saposins/pharmacology , alpha-Synuclein/metabolism
6.
PLoS One ; 10(5): e0126856, 2015.
Article in English | MEDLINE | ID: mdl-25993033

ABSTRACT

Four sphingolipid activator proteins (i.e., saposins A-D) are synthesized from a single precursor protein, prosaposin (PS), which exerts exogenous neurotrophic effects in vivo and in vitro. Kainic acid (KA) injection in rodents is a good model in which to study neurotrophic factor elevation; PS and its mRNA are increased in neurons and the choroid plexus in this animal model. An 18-mer peptide (LSELIINNATEELLIKGL; PS18) derived from the PS neurotrophic region prevents neuronal damage after ischemia, and PS18 is a potent candidate molecule for use in alleviating ischemia-induced learning disabilities and neuronal loss. KA is a glutamate analog that stimulates excitatory neurotransmitter release and induces ischemia-like neuronal degeneration; it has been used to define mechanisms involved in neurodegeneration and neuroprotection. In the present study, we demonstrate that a subcutaneous injection of 0.2 and 2.0 mg/kg PS18 significantly improved behavioral deficits of Wistar rats (n = 6 per group), and enhanced the survival of hippocampal and cortical neurons against neurotoxicity induced by 12 mg/kg KA compared with control animals. PS18 significantly protected hippocampal synapses against KA-induced destruction. To evaluate the extent of PS18- and KA-induced effects in these hippocampal regions, we performed histological evaluations using semithin sections stained with toluidine blue, as well as ordinal sections stained with hematoxylin and eosin. We revealed a distinctive feature of KA-induced brain injury, which reportedly mimics ischemia, but affects a much wider area than ischemia-induced injury: KA induced neuronal degeneration not only in the CA1 region, where neurons degenerate following ischemia, but also in the CA2, CA3, and CA4 hippocampal regions.


Subject(s)
Brain Injuries/drug therapy , Saposins/pharmacology , Amino Acid Sequence , Animals , Avoidance Learning/drug effects , Brain Injuries/chemically induced , Brain Injuries/pathology , Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Kainic Acid/toxicity , Male , Molecular Sequence Data , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Neurons/drug effects , Neurons/pathology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Saposins/chemistry , Saposins/genetics , Synapses/drug effects , Synapses/pathology
7.
Mol Cancer Ther ; 14(2): 491-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25670331

ABSTRACT

Lung cancer is the deadliest type of cancer for both men and women. In this study, we evaluate the in vitro and in vivo efficacy of a biotherapeutic agent composed of a lysosomal protein (Saposin C, SapC) and a phospholipid (dioleoylphosphatidylserine, DOPS), which can be assembled into nanovesicles (SapC-DOPS) with selective antitumor activity. SapC-DOPS targets phosphatidylserine, an anionic phospholipid preferentially exposed in the surface of cancer cells and tumor-associated vasculature. Because binding of SapC to phosphatidylserine is favored at acidic pHs, and the latter characterizes the milieu of many solid tumors, we tested the effect of pH on the binding capacity of SapC-DOPS to lung tumor cells. Results showed that SapC-DOPS binding to cancer cells was more pronounced at low pH. Viability assays on a panel of human lung tumor cells showed that SapC-DOPS cytotoxicity was positively correlated with cell surface phosphatidylserine levels, whereas mitochondrial membrane potential measurements were consistent with apoptosis-related cell death. Using a fluorescence tracking method in live mice, we show that SapC-DOPS specifically targets human lung cancer xenografts, and that systemic therapy with SapC-DOPS induces tumor apoptosis and significantly inhibits tumor growth. These results suggest that SapC-DOPS nanovesicles are a promising treatment option for lung cancer.


Subject(s)
Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Nanostructures/chemistry , Phosphatidylserines/chemistry , Saposins/therapeutic use , Unilamellar Liposomes/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Mice, Nude , Saposins/pharmacology , Xenograft Model Antitumor Assays
8.
Oncotarget ; 5(20): 9703-9, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25210852

ABSTRACT

SapC-DOPS is a novel nanotherapeutic that has been shown to target and induce cell death in a variety of cancers, including glioblastoma (GBM). GBM is a primary brain tumor known to frequently demonstrate resistance to apoptosis-inducing therapeutics. Here we explore the mode of action for SapC-DOPS in GBM, a treatment being developed by Bexion Pharmaceuticals for clinical testing in patients. SapC-DOPS treatment was observed to induce lysosomal dysfunction of GBM cells characterized by decreased glycosylation of LAMP1 and altered proteolytic processing of cathepsin D independent of apoptosis and autophagic cell death. We observed that SapC-DOPS induced lysosomal membrane permeability (LMP) as shown by LysoTracker Red and Acridine Orange staining along with an increase of sphingosine, a known inducer of LMP. Additionally, SapC-DOPS displayed strong synergistic interactions with the apoptosis-inducing agent TMZ. Collectively our data suggest that SapC-DOPS induces lysosomal cell death in GBM cells, providing a new approach for treating tumors resistant to traditional apoptosis-inducing agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Nanostructures/administration & dosage , Phosphatidylserines/pharmacology , Saposins/pharmacology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Lysosomes/drug effects , Mice , Mice, Nude , Random Allocation , Saposins/administration & dosage , Temozolomide , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 110(23): 9529-34, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690594

ABSTRACT

GPR37 (also known as Pael-R) and GPR37L1 are orphan G protein-coupled receptors that are almost exclusively expressed in the nervous system. We screened these receptors for potential activation by various orphan neuropeptides, and these screens yielded a single positive hit: prosaptide, which promoted the endocytosis of GPR37 and GPR37L1, bound to both receptors and activated signaling in a GPR37- and GPR37L1-dependent manner. Prosaptide stimulation of cells transfected with GPR37 or GPR37L1 induced the phosphorylation of ERK in a pertussis toxin-sensitive manner, stimulated (35)S-GTPγS binding, and promoted the inhibition of forskolin-stimulated cAMP production. Because prosaptide is the active fragment of the secreted neuroprotective and glioprotective factor prosaposin (also known as sulfated glycoprotein-1), we purified full-length prosaposin and found that it also stimulated GPR37 and GPR37L1 signaling. Moreover, both prosaptide and prosaposin were found to protect primary astrocytes against oxidative stress, with these protective effects being attenuated by siRNA-mediated knockdown of endogenous astrocytic GPR37 or GPR37L1. These data reveal that GPR37 and GPR37L1 are receptors for the neuroprotective and glioprotective factors prosaptide and prosaposin.


Subject(s)
Nerve Growth Factors/metabolism , Receptors, G-Protein-Coupled/metabolism , Saposins/metabolism , Animals , Astrocytes/drug effects , Blotting, Western , COS Cells , Chlorocebus aethiops , Cyclic AMP/biosynthesis , Gene Knockdown Techniques , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nerve Growth Factors/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Polysorbates , RNA, Small Interfering/genetics , Saposins/pharmacology , Sulfur Radioisotopes/metabolism
10.
Neuroscience ; 236: 373-93, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23321539

ABSTRACT

Parkinson's disease (PD) is a chronic progressive neurological disorder with an increasing incidence in the aging population. Neuroprotective and/or neuroregenerative strategies remain critical in the treatment of this increasingly prevalent disease. Prosaposin is a neurotrophic factor whose neurotrophic activity is attributed to a stretch of 12 amino acids located at the N-terminal region of saposin C. The present study was performed to investigate the protective effect and mechanism of action of a prosaposin-derived 18-mer peptide (PS18: LSELIINNATEELLIKGL) in Parkinson's disease models. We used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium ion (MPP(+))-induced dopaminergic neurotoxicity in C57BL/6J mice or SH-SY5Y cells and explored the protective effect and mechanisms of action of PS18 on dopaminergic neurons. Treatment with 2.0mg/kg PS18 significantly improved behavioral deficits, enhanced the survival of tyrosine hydroxylase-positive neurons, and decreased the activity of astrocytes in the substantia nigra and striatum in MPTP-induced PD model mice. In vitro, a Cell Counting Kit-8 assay and Hoechst 33258 staining revealed that co-treatment with 300ng/mL PS18 and 5mM MPP(+) protected against MPP(+)-induced nuclear morphological changes and attenuated cell death induced by MPP(+). We also found that PS18-FAM entered the cells, and the retention time of PS18-FAM in the cytoplasm of MPP(+)-treated cells was shorter than that of untreated cells. In addition, PS18 showed protection from MPP(+)/MPTP-induced apoptosis in the SH-SY5Y cells and dopaminergic neurons in the PD model mice via suppression of the c-Jun N-terminal kinase/c-Jun pathway; upregulation of Bcl-2; downregulation of BAX, attenuating mitochondrial damage; and inhibition of caspase-3. These findings suggest that PS18 may provide a valuable therapeutic strategy for the treatment of progressive neurodegenerative diseases such as PD.


Subject(s)
Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Saposins/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology
11.
Biochem J ; 441(2): 665-74, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21943334

ABSTRACT

TFPI-2 (tissue factor pathway inhibitor-2) has recently been recognized as a new tumour suppressor gene. Low expression of this protein in several types of cancers allows for enhanced tumour growth, invasion and metastasis. To investigate the molecular mechanism responsible for the tumour-suppressor effects of TFPI-2, we performed yeast two-hybrid analysis and identified PSAP (prosaposin) as a TFPI-2-interacting partner. This interaction was confirmed by co-immunoprecipitation and immunofluorescence. The region of TFPI-2 that interacts with PSAP is located in the KD2 (Kunitz-type domain 2). Further study showed that PSAP does not affect the function of TFPI-2 as a serine proteinase inhibitor, but that TFPI-2 could inhibit the invasion-promoting effects of PSAP in human HT1080 fibrosarcoma cells. The results of the present study revealed that TFPI-2 interacts with PSAP, which may play an important role in the physiology and pathology of diseases such as cancer.


Subject(s)
Cell Movement/ethics , Fibrosarcoma/physiopathology , Glycoproteins/metabolism , Neoplasm Invasiveness , Saposins/metabolism , Animals , Binding Sites , COS Cells , Cell Movement/drug effects , Chlorocebus aethiops , HEK293 Cells , Humans , Matrix Metalloproteinases/metabolism , Protein Structure, Tertiary , Saposins/pharmacology , Serine Proteinase Inhibitors/genetics , Two-Hybrid System Techniques
12.
Inflamm Res ; 60(2): 153-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20853174

ABSTRACT

OBJECTIVE AND DESIGN: SapC-DOPS is a newly combined compound consisting of saposin C and dioleoylphosphatidylserine (DOPS). Our recent study showed that SapC-DOPS exhibits anti-tumor activity. However, SapC-DOPS has recognition elements of Toll-like receptor (TLR) 2 and TLR4; therefore, we want to know whether SapC-DOPS can induce abnormal immunoreaction via identification TLRs. METHODS: We investigated the capacity of SapC-DOPS to induce cytokines in vivo and in vitro and analyzed the involvement of TLR and NF-kB in these cytokines production. RESULTS: SapC-DOPS could activate the cytokine production by peripheral macrophages, enhance the expressions of TLR4 and stimulate the NF-κB nuclear translocation. PDTC, an NF-κB inhibitor, could decrease the SapC-DOPS inducible TNF-α and IL-1ß production. CONCLUSIONS: SapC-DOPS was similar to LPS in the immune response and may induce the production of cytokines in macrophages via the TLR4 signaling pathway and, at least in part, the alteration of the NF-κB pathway.


Subject(s)
Cytokines/immunology , Macrophages/drug effects , Macrophages/immunology , Phosphatidylserines/pharmacology , Saposins/pharmacology , Toll-Like Receptor 4/immunology , Animals , Cell Line , Female , Macrophages/cytology , Mice , Mice, Inbred C57BL , Phosphatidylserines/chemistry , Phosphatidylserines/immunology , Saposins/chemistry , Saposins/immunology , Signal Transduction/immunology
13.
J Neurochem ; 112(6): 1527-38, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20050969

ABSTRACT

Functionally, adult stem cells not only participate in replication and differentiation to various cell lineages, but also may be involved in rescuing cells from apoptosis. Identifying functional factors secreted by stem cells, as well as their target cells, may advance our understanding of stem cells' multifaceted physiologic functions. Here, we report that mouse bone marrow stromal cell-derived neuroprogenitor cells (mMSC-NPC) provide a protective function by secreting a key factor, prosaposin (PSAP), capable of rescuing mature neurons from apoptotic death. This factor is identified as the lead protein in the secretome of mMSC-NPC cultures by tandem mass spectroscopic profiling, and further validated by western blotting and immunocytochemistry. The secretome of MSC-NPC reduces toxin-induced cell death in cultures of rat pheochromocytoma neuronal cells, human ReNcell CX neurons, and rat cortical primary neurons; removal of PSAP by immunodepletion annuls this protective effect. This neuronal protection against toxin treatment was validated further by the recombinant PSAP peptide. Interestingly, the secretome of neuronal culture does not possess such a self-protective action. We suggest that upon injury, a subgroup of MSCs differentiates into neural/neuronal progenitor cells, and remains in this intermediate stem cell-like stage, defending injured neighboring mature neurons from apoptosis by secreting PSAP.


Subject(s)
Apoptosis/drug effects , Bone Marrow Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Saposins/metabolism , Saposins/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Annexin A5/metabolism , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Chromatography, High Pressure Liquid/methods , Culture Media, Conditioned/chemistry , Humans , Mice , Propidium , Rats , Tretinoin/pharmacology
14.
Clin Cancer Res ; 15(18): 5840-51, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19737950

ABSTRACT

PURPOSE: Saposin C is a multifunctional protein known to activate lysosomal enzymes and induce membrane fusion in an acidic environment. Excessive accumulation of lipid-coupled saposin C in lysosomes is cytotoxic. Because neoplasms generate an acidic microenvironment, caused by leakage of lysosomal enzymes and hypoxia, we hypothesized that saposin C may be an effective anticancer agent. We investigated the antitumor efficacy and systemic biodistribution of nanovesicles comprised of saposin C coupled with dioleoylphosphatidylserine in preclinical cancer models. EXPERIMENTAL DESIGN: Neuroblastoma, malignant peripheral nerve sheath tumor and, breast cancer cells were treated with saposin C-dioleoylphosphatidylserine nanovesicles and assessed for cell viability, ceramide elevation, caspase activation, and apoptosis. Fluorescently labeled saposin C-dioleoylphosphatidylserine was i.v. injected to determine in vivo tumor-targeting specificity. Antitumor activity and toxicity profile of saposin C-dioleoylphosphatidylserine were evaluated in xenograft models. RESULTS: Saposin C-dioleoylphosphatidylserine nanovesicles, with a mean diameter of approximately 190 nm, showed specific tumor-targeting activity shown through in vivo imaging. Following i.v. administration, saposin C-dioleoylphosphatidylserine nanovesicles preferentially accumulated in tumor vessels and cells in tumor-bearing mice. Saposin C-dioleoylphosphatidylserine induced apoptosis in multiple cancer cell types while sparing normal cells and tissues. The mechanism of saposin C-dioleoylphosphatidylserine induction of apoptosis was determined to be in part through elevation of intracellular ceramides, followed by caspase activation. In in vivo models, saposin C-dioleoylphosphatidylserine nanovesicles significantly inhibited growth of preclinical xenografts of neuroblastoma and malignant peripheral nerve sheath tumor. I.v. dosing of saposin C-dioleoylphosphatidylserine showed no toxic effects in nontumor tissues. CONCLUSIONS: Saposin C-dioleoylphosphatidylserine nanovesicles offer promise as a novel, nontoxic, cancer-targeted, antitumor agent for treating a broad range of cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lysosomes/chemistry , Neoplasms/drug therapy , Phosphatidylserines/chemistry , Saposins/pharmacology , Saposins/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Liposomes , Mice , Neoplasms/pathology , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/pathology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Saposins/chemistry , Saposins/metabolism , Substrate Specificity , Tumor Cells, Cultured
15.
Proc Natl Acad Sci U S A ; 104(44): 17394-9, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17954913

ABSTRACT

Acid beta-glucosidase (GCase) is a soluble lysosomal enzyme responsible for the hydrolysis of glucose from glucosylceramide and requires activation by the small nonenzymatic protein saposin C (sapC) to gain access to the membrane-embedded glycosphingolipid substrate. We have used in situ atomic force microscopy (AFM) with simultaneous confocal and epifluorescence microscopies to investigate the interactions of GCase and sapC with lipid bilayers. GCase binds to sites on membranes transformed by sapC, and enzyme activity occurs at loci containing both GCase and sapC. Using FRET, we establish the presence of GCase/sapC and GCase/product contacts in the bilayer. These data support a mechanism in which sapC locally alters regions of bilayer for subsequent attack by the enzyme in stably bound protein complexes.


Subject(s)
Lipid Bilayers/metabolism , Saposins/pharmacology , beta-Glucosidase/metabolism , Enzyme Activation/drug effects , Microscopy, Atomic Force , Protein Binding , Saposins/genetics , Saposins/metabolism
16.
Prostate ; 67(2): 178-89, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17044040

ABSTRACT

BACKGROUND: Prosaposin overexpression and/or genomic amplification have been demonstrated in androgen-independent (AI) prostate cancer cell lines and tissues. Here, we explored the possibility for a functional relationship between prosaposin and androgen receptor (AR) in LNCaP cells. METHODS: The effect of prosaposin or its active molecular derivatives (e.g., saposin C) on expression and activity of androgen receptor (AR) and prostate-specific antigen (PSA) was examined by using immunoblotting, RT-PCR, transfection, and reporter gene assays, immunofluorescence staining, and inhibitors of signal transduction pathways. RESULTS: Prosaposin or saposin C, in an AI-manner, (a) increased AR mRNA and protein expression and nuclear AR content and its phosphorylation state; (b) increased PSA mRNA and protein expression; and (c) upregulated PSA- and an androgen-inducible probasin (PB)-reporter gene activity in LNCaP and AR-transfected PC-3 cells. Induction of PSA expression and reporter activity was substantially blocked or prevented with the antiandrogen bicalutamide, pertussis toxin, or inhibitors of MAPK- and PI3K/Akt-signaling pathways, indicating an androgen-agonistic effect for saposin C that involves AR and multiple signaling pathways. CONCLUSIONS: The results for the first time introduce prosaposin as an androgen-agonist in prostate cancer cells. This finding, together with the growth-promoting effect and overexpression of prosaposin, may support a growth advantage to AI prostate cancer cells.


Subject(s)
Prostate-Specific Antigen/genetics , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Saposins/pharmacology , Androgen Antagonists/pharmacology , Androgen-Binding Protein/genetics , Androgen-Binding Protein/metabolism , Anilides/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , Male , Nitriles , Plasmids , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tosyl Compounds , Transfection , Up-Regulation/drug effects
17.
Neurosci Res ; 56(4): 372-85, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17050025

ABSTRACT

Neurotrophic factors are a group of secreted proteins which generally regulate neurite outgrowth and synaptic development. SGP-1 has been reported as a neurotrophic factor, though little is known of its effect on neurite outgrowth, and it is unknown whether SGP-1 affects synaptic development. We report here that SGP-1 is distributed in vesicle-like puncta in somas and dendrites of primary neurons in culture, and that SGP-1 is secreted in culture and is taken up by endocytosis in dendrites. Endogenous extracellular activity of SGP-1 promotes dendritic, but not axonal outgrowth. Furthermore, endogenous activity of SGP-1 increases synaptogenesis in hippocampal neurons as determined by measuring the density and size of synaptophysin puncta and by determining the density of dendritic spines, their surface expression of GluR2 and their immunoreactivity for GluR1. The effect of SGP-1 on the amount of postsynaptic receptors in dendritic spines depends on synaptic activity and apparently on activation of MAPK, as inhibition of either of these abolished the affect. Hence, SGP-1 has neurotrophic effects, increasing dendritic growth and promoting synaptic development in an activity-dependent fashion.


Subject(s)
Dendrites/drug effects , Saposins/pharmacology , Synapses/drug effects , Animals , Axons/drug effects , Cells, Cultured , Endocytosis/drug effects , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Hippocampus/cytology , Hippocampus/drug effects , Immunoglobulin G/immunology , Immunohistochemistry , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Transfection
18.
Mol Biochem Parasitol ; 147(1): 85-94, 2006 May.
Article in English | MEDLINE | ID: mdl-16529828

ABSTRACT

Amoebapores, the pore-forming proteins of Entamoeba histolytica, are major pathogenicity factors of the parasite. Upon a comprehensive survey in the recently completed genome data sets for the protozoon, we identified in addition to the three amoebapore genes, 16 genes which are constitutively expressed and code for structurally similar proteins, all belonging to the family of saposin-like proteins. Here, we recombinantly expressed in bacteria a defined single entity of this expansive amoebic protein family, namely SAPLIP 3. The protein consists of the saposin-like domain only, comparable to amoebapores, and we characterized its interactions with membranes using different assays. In contrast to amoebapores, SAPLIP 3 neither forms pores in liposomes nor permeabilizes bacterial membranes. However, SAPLIP 3 induces leaky fusion of lipid vesicles as evidenced by fluorescence microscopic analysis and by using a fusion assay that monitors the dequenching of a lipophilic dye. The membrane-fusogenic activity of SAPLIP 3 which is dependent on the presence of negatively charged lipids and on acidic pH resembles in combination with the negative surface charge of the protein characteristics of human saposin C. Beside its function as a cofactor of sphingolipid hydrolysing enzymes, the human protein is considered to be involved in the reorganization of lysosomal compartments due to its fusogenic activity. We hypothesize that in the amoeba, SAPLIP 3 fulfils a similar function in the multifarious endo- and exocytotic transport processes.


Subject(s)
Entamoeba histolytica/physiology , Membrane Fusion , Protozoan Proteins/metabolism , Saposins/metabolism , Animals , Endocytosis , Entamoeba histolytica/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Ion Channels/metabolism , Liposomes/metabolism , Microbial Sensitivity Tests , Models, Molecular , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/pharmacology , Saposins/chemistry , Saposins/genetics , Saposins/pharmacology
19.
Behav Neurosci ; 120(1): 1-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16492111

ABSTRACT

Estrogens modulate almost all aspects of female behavioral arousal; however, apart from that of sexual behavior, the neurobiology of female arousal remains unclear. Because orexins-hypocretins are neurotransmitters known to be important for behavioral arousal, the authors hypothesized that orexins may be a target for estrogen. Gonadectomized female mice received an intracerebral injection of either phosphate-buffered saline, the neurotoxin saporin (SAP), or the orexin-2-saporin conjugate (OXSAP) in the lateral hypothalamus. SAP- and OXSAP-treated mice were also divided into groups receiving either estradiol capsules or oil capsules. Mice were tested in 3 behavioral tests measuring different modes of arousal: sensory responsiveness, running wheel activity, and fearfulness. OXSAP mice showed decreases in sensory responsiveness and fearfulness concomitant with a reduction in orexin cell number. Estradiol affected all behaviors tested but decreased fearfulness only when combined with OXSAP treatment. These data indicate that estrogens modulate orexins' effects on fearfulness.


Subject(s)
Arousal/drug effects , Behavior, Animal/drug effects , Brain/cytology , Brain/drug effects , Estradiol/pharmacology , Fear/drug effects , Neuropeptides/pharmacology , Saposins/pharmacology , Animals , Estradiol/administration & dosage , Female , Injections , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Neuropeptides/administration & dosage , Orexins , Pilot Projects , Saposins/administration & dosage
20.
J Biol Chem ; 281(7): 4242-53, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16293621

ABSTRACT

Acid beta-glucosidase (GCase) is a 497-amino acid, membrane-associated lysosomal exo-beta-glucosidase whose defective activity leads to the Gaucher disease phenotypes. To move toward a structure/function map for disease mutations, 52 selected single amino acid substitutions were introduced into GCase, expressed in an insect cell system, purified, and characterized for basic kinetic, stability, and activator response properties. The variant GCases from Gaucher disease patients and selected variant GCases from the mouse had decreased relative k(cat) and differential effects on active site binding and/or attachment of mechanism-based covalent (conduritol B epoxide) or reversible (deoxynojirimycin derivatives) inhibitors. A defect in negatively charged phospholipid activation was present in the majority of variant GCases but was increased in two, N370S and V394L. Deficits in saposin C enhancement of k(cat) were present in variant GCases involving residues 48-122, whereas approximately 2-fold increases were obtained with the L264I GCase. About 50% of variant GCases each had wild-type or increased sensitivity to in vitro cathepsin D digestion. Mapping of these properties onto the crystal structures of GCase indicated wide dispersion of functional properties that can affect catalytic function and stability. Site-directed mutagenesis of cysteine residues showed that the disulfide bonds, Cys(4)-Cys(16) and Cys(18)-Cys(23), and a free Cys(342) were essential for activity; the free Cys(126) and Cys(248) were not. Relative k(cat) was highly sensitive to a His substitution at Arg(496) but not at Arg(495). These studies and high phylogenetic conservation indicate localized and general structural effects of Gaucher disease mutations that were not obvious from the nature of the amino acid substitution, including those predicted to be nondisruptive (e.g. Val --> Leu). These results provide initial studies for the engineering of variant GCases and, potentially, molecular chaperones for therapeutic use.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Point Mutation , Amino Acid Sequence , Crystallization , Enzyme Activation , Glucosylceramidase/chemistry , Glycosylation , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Phosphatidylserines/pharmacology , Saposins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...