Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 539: 215698, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35523311

ABSTRACT

Macrophages play important roles in cancer microenvironment. Human cytosolic glycyl-tRNA synthetase (GARS1) was previously shown to be secreted via extracellular vesicles (EVs) from macrophages to trigger cancer cell death. However, the effects of GARS1-containing EVs (GARS1-EVs) on macrophages as well as on cancer cells and the working mechanisms of GARS1 in cancer microenvironment are not yet understood. Here we show that GARS1-EVs induce M1 polarization and facilitate phagocytosis of macrophages. GARS1-EVs triggers M1 polarization of macrophage via the specific interaction of the extracellular cadherin subdomains 1-4 of the cadherin EGF LAG seven-pass G-type receptor 2 (CELSR2) with the N-terminal WHEP domain containing peptide region of GARS1, and activates the RAF-MEK-ERK pathway for M1 type cytokine production and phagocytosis. Besides, GARS1 interacted with cadherin 6 (CDH6) of cancer cells via its C-terminal tRNA-binding domain to induce cancer cell death. In vivo model, GARS1-EVs showed potent suppressive activity against tumor initiation via M1 type macrophages. GARS1 displayed on macrophage-secreted extracellular vesicles suppressed tumor growth in dual mode, namely through pro-apoptotic effect on cancer cells and M1 polarization effect on macrophages. Collectively, these results elucidate the unique tumor suppressive activity and mechanism of GARS1-EVs by activating M1 macrophage via CELSR2 as well as by direct killing of cancer cells via CDH6.


Subject(s)
Extracellular Vesicles , Glycine-tRNA Ligase , Macrophages , Neoplasms , Cadherins/metabolism , Cell Polarity , Extracellular Vesicles/enzymology , Extracellular Vesicles/metabolism , Glycine-tRNA Ligase/analysis , Glycine-tRNA Ligase/metabolism , Glycine-tRNA Ligase/pharmacology , Humans , Macrophages/enzymology , Macrophages/metabolism , Macrophages/pathology , Neoplasms/enzymology , Neoplasms/metabolism , Phagocytosis , Tumor Microenvironment
2.
Future Med Chem ; 14(11): 827-845, 2022 06.
Article in English | MEDLINE | ID: mdl-35502655

ABSTRACT

Extracellular vesicles (EVs) are nanovesicles secreted for intercellular communication with endosomal network regulating secretion of small EVs (or exosomes) that play roles in cancer progression. As an essential oncoprotein, Kirsten rat sarcoma virus (KRAS) is tightly regulated by its endosomal trafficking for membrane attachment. However, the crosstalk between KRAS and EVs has been scarcely discussed despite its endocytic association. An overview of the oncogenic role of KRAS focusing on its correlation with cancer-associated EVs should provide important clues for disease prognosis and inspire novel therapeutic approaches for treating KRAS mutant cancers. Therefore, this review summarizes the relevant studies that provide substantial evidence linking KRAS mutation to EVs and discusses the oncogenic implication from the aspects of biogenesis, cargo sorting, and release and uptake of the EVs.


Subject(s)
Extracellular Vesicles , Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Biological Transport , Carcinogenesis/metabolism , Cell Communication , Extracellular Vesicles/enzymology , Extracellular Vesicles/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism
3.
Clin Pharmacol Ther ; 111(2): 425-434, 2022 02.
Article in English | MEDLINE | ID: mdl-34623637

ABSTRACT

Preparations of plasma-derived small extracellular vesicles (sEVs) were deployed as liquid biopsy to study cytochrome P450 (CYP) 3A4 (CYP3A4) induction following modafinil 400 mg once daily × 14 days (young healthy volunteers, N = 10 subjects). Induction was confirmed using the 4ß-hydroxycholesterol-to-cholesterol (4ßHC/C) ratio, a plasma CYP3A4/5 biomarker, with a mean 2.1-fold increase (Day 15 vs. Day 1; 90% confidence interval (CI) = 1.8-2.3; P value = 0.0004). Proteomic analysis revealed the induction (mean Day 15 vs. Day 1 fold-increase (90% CI)) of both liver (1.3 (1.1-1.5), P value = 0.014) and nonliver (1.9 (1.6-2.2), P value = 0.04) sEV CYP3A4 protein expression. In CYP3A5 nonexpresser subjects, the baseline (pre-dose) 4ßHC/C plasma ratio was more highly correlated with liver sEVs (r = 0.937, P value = 0.001) than nonliver sEVs (r = 0.619, P value = 0.101) CYP3A4 protein expression. When CYP3A5 expressers (CYP3A5*1/*3) were included, the correlation with liver sEVs (r = 0.761, P value = 0.011) and nonliver sEVs (r = 0.391, P value = 0.264) CYP3A4 protein was weaker. Although modafinil-induced changes in plasma 4ßHC/C ratio did not correlate with sEVs CYP3A4 protein expression, the individual subject sEVs proteomic data were used successfully to predict victim drug (midazolam, triazolam, dextromethorphan, 17α-ethinylestradiol, and abemaciclib) area under the plasma concentration-time curve (AUC) ratios (AUCRs) following modafinil. Based on the AUCR values, modafinil was classified as a weak to moderate CYP3A4 inducer (vs. rifampicin). For the first time, it was possible to deploy plasma-derived sEVs to study CYP3A4 induction beyond rifampicin, a more potent CYP3A4 inducer.


Subject(s)
Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A/biosynthesis , Modafinil/administration & dosage , Biomarkers/blood , Cytochrome P-450 CYP3A/blood , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A Inducers/adverse effects , Drug Administration Schedule , Drug Interactions , Enzyme Induction , Extracellular Vesicles/drug effects , Extracellular Vesicles/enzymology , Healthy Volunteers , Humans , Hydroxycholesterols/blood , Liquid Biopsy , Liver/enzymology , Modafinil/adverse effects , Models, Biological , Plasma/enzymology , Proteomics , Rifampin/administration & dosage , Rifampin/adverse effects , Time Factors
4.
Open Biol ; 11(12): 210276, 2021 12.
Article in English | MEDLINE | ID: mdl-34847775

ABSTRACT

Amplification of the proto-oncogene MYCN is a key molecular aberration in high-risk neuroblastoma and predictive of poor outcome in this childhood malignancy. We investigated the role of MYCN in regulating the protein cargo of extracellular vesicles (EVs) secreted by tumour cells that can be internalized by recipient cells with functional consequences. Using a switchable MYCN system coupled to mass spectrometry analysis, we found that MYCN regulates distinct sets of proteins in the EVs secreted by neuroblastoma cells. EVs produced by MYCN-expressing cells or isolated from neuroblastoma patients induced the Warburg effect, proliferation and c-MYC expression in target cells. Mechanistically, we linked the cancer-promoting activity of EVs to the glycolytic kinase pyruvate kinase M2 (PKM2) that was enriched in EVs secreted by MYC-expressing neuroblastoma cells. Importantly, the glycolytic enzymes PKM2 and hexokinase II were detected in the EVs circulating in the bloodstream of neuroblastoma patients, but not in those of non-cancer children. We conclude that MYC-activated cancers might spread oncogenic signals to remote body locations through EVs.


Subject(s)
Carrier Proteins/metabolism , Extracellular Vesicles/enzymology , Hexokinase/metabolism , Membrane Proteins/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Proteomics/methods , Thyroid Hormones/metabolism , Carrier Proteins/blood , Cell Line, Tumor , Cell Proliferation , Child , Gene Amplification , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glycolysis , Hexokinase/blood , Humans , Mass Spectrometry , Membrane Proteins/blood , Neuroblastoma/blood , Phosphorylation , Thyroid Hormones/blood , Thyroid Hormone-Binding Proteins
5.
Cells ; 10(11)2021 10 30.
Article in English | MEDLINE | ID: mdl-34831185

ABSTRACT

Phospholipids are major components in the lipid bilayer of cell membranes. These molecules are comprised of two acyl or alkyl groups and different phospho-base groups linked to the glycerol backbone. Over the years, substantial interest has focused on metabolism of phospholipids by phospholipases and the role of their metabolic products in mediating cell functions. The high levels of polyunsaturated fatty acids (PUFA) in the central nervous system (CNS) have led to studies centered on phospholipases A2 (PLA2s), enzymes responsible for cleaving the acyl groups at the sn-2 position of the phospholipids and resulting in production of PUFA and lysophospholipids. Among the many subtypes of PLA2s, studies have centered on three major types of PLA2s, namely, the calcium-dependent cytosolic cPLA2, the calcium-independent iPLA2 and the secretory sPLA2. These PLA2s are different in their molecular structures, cellular localization and, thus, production of lipid mediators with diverse functions. In the past, studies on specific role of PLA2 on cells in the CNS are limited, partly because of the complex cellular make-up of the nervous tissue. However, understanding of the molecular actions of these PLA2s have improved with recent advances in techniques for separation and isolation of specific cell types in the brain tissue as well as development of sensitive molecular tools for analyses of proteins and lipids. A major goal here is to summarize recent studies on the characteristics and dynamic roles of the three major types of PLA2s and their oxidative products towards brain health and neurological disorders.


Subject(s)
Central Nervous System Diseases/enzymology , Central Nervous System Diseases/pathology , Central Nervous System/enzymology , Central Nervous System/pathology , Phospholipases A2, Secretory/metabolism , Extracellular Vesicles/enzymology , Humans , Lipid Peroxidation , Lipidomics , Phospholipases A2, Secretory/chemistry
6.
Biochem Pharmacol ; 194: 114796, 2021 12.
Article in English | MEDLINE | ID: mdl-34678224

ABSTRACT

Extracellular Vesicles (EVs) are implicated in the spread of pathogenic proteinsin a growing number of neurological diseases. Given this, there is rising interest in developing inhibitors of Neutral Sphingomyelinase 2 (nSMase2), an enzyme critical in EV biogenesis. Our group recently discovered phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate (PDDC), the first potent, selective, orally-available, and brain-penetrable nSMase2 inhibitor, capable of dose-dependently reducing EVs release in vitro and in vivo. Herein, using multiplexed Surface Plasmon Resonance imaging (SPRi), we evaluated which brain cell-derived EVs were affected by PDDC following acute brain injury. Mice were fed PDDC-containing chow at doses which gave steady PDDC brain exposures exceeding its nSMase2 IC50. Mice were then administered an intra-striatal IL-1ß injection and two hours later plasma and brain were collected. IL-1ß injection significantly increased striatal nSMase2 activity which was completely normalized by PDDC. Using SPRi, we found that IL-1ß-induced injury selectively increased plasma levels of CD171 + and PLP1 + EVs; this EV increase was normalized by PDDC. In contrast, GLAST1 + EVs were unchanged by IL-1ß or PDDC. IL-1ß injection selectively increased EVs released from activated versus non-activated microglia, indicated by the CD11b+/IB4 + ratio. The increase in EVs from CD11b + microglia was dramatically attenuated with PDDC. Taken together, our data demonstrate that following acute injury, brain nSMase2 activity is elevated. EVs released from neurons, oligodendrocytes, and activated microglial are increased in plasma and inhibition of nSMase2 with PDDC reduced these IL-1ß-induced changes implicating nSMase2 inhibition as a therapeutic target for acute brain injury.


Subject(s)
Brain Injuries/enzymology , Extracellular Vesicles/enzymology , Microglia/enzymology , Neurons/enzymology , Oligodendroglia/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Brain Injuries/drug therapy , Carnitine/administration & dosage , Carnitine/analogs & derivatives , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Extracellular Vesicles/drug effects , Injections, Intraventricular , Interleukin-1beta/administration & dosage , Male , Mice , Mice, Transgenic , Microglia/drug effects , Neurons/drug effects , Oligodendroglia/drug effects , Pyrenes/administration & dosage , Sphingomyelin Phosphodiesterase/antagonists & inhibitors
7.
J Biol Chem ; 297(4): 101198, 2021 10.
Article in English | MEDLINE | ID: mdl-34534548

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) fulfills various physiological roles that are unrelated to its glycolytic function. However, to date, the nonglycolytic function of GAPDH in trypanosomal parasites is absent from the literature. Exosomes secreted from Leishmania, like entire parasites, were found to have a significant impact on macrophage cell signaling and function, indicating cross talk with the host immune system. In this study, we demonstrate that the Leishmania GAPDH (LmGAPDH) protein is highly enriched within the extracellular vesicles (EVs) secreted during infection. To understand the function of LmGAPDH in EVs, we generated control, overexpressed, half-knockout (HKO), and complement cell lines. HKO cells displayed lower virulence compared with control cells when macrophages and BALB/c mice were infected with them, implying a crucial role for LmGAPDH in Leishmania infection and disease progression. Furthermore, upon infection of macrophages with HKO mutant Leishmania and its EVs, despite no differences in TNFA mRNA expression, there was a considerable increase in TNF-α protein expression compared with control, overexpressed, and complement parasites as determined by ELISA, RT-PCR, and immunoblot data. In vitro protein translation studies suggest that LmGAPDH-mediated TNF-α suppression occurs in a concentration-dependent manner. Moreover, mRNA binding assays also verified that LmGAPDH binds to the AU-rich 3'-UTR region of TNFA mRNA, limiting its production. Together, these findings confirmed that the LmGAPDH contained in EVs inhibits TNF-α expression in macrophages during infection via posttranscriptional repression.


Subject(s)
Extracellular Vesicles/enzymology , Gene Expression Regulation , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Leishmania major/enzymology , Macrophages/metabolism , Protozoan Proteins/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Extracellular Vesicles/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/immunology , Leishmania major/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Tumor Necrosis Factor-alpha/immunology
8.
Am J Physiol Renal Physiol ; 321(5): F659-F673, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34569252

ABSTRACT

Since previous research suggests a role of a circulating factor in the pathogenesis of steroid-sensitive nephrotic syndrome (NS), we speculated that circulating plasma extracellular vesicles (EVs) are a candidate source of such a soluble mediator. Here, we aimed to characterize and try to delineate the effects of these EVs in vitro. Plasma EVs from 20 children with steroid-sensitive NS in relapse and remission, 10 healthy controls, and 6 disease controls were obtained by serial ultracentrifugation. Characterization of these EVs was performed by electron microscopy, flow cytometry, and Western blot analysis. Major proteins from plasma EVs were identified via mass spectrometry. Gene Ontology classification analysis and Ingenuity Pathway Analysis were performed on selectively expressed EV proteins during relapse. Immortalized human podocyte culture was used to detect the effects of EVs on podocytes. The protein content and particle number of plasma EVs were significantly increased during NS relapse. Relapse NS EVs selectively expressed proteins that involved actin cytoskeleton rearrangement. Among these, the level of RAC-GTP was significantly increased in relapse EVs compared with remission and disease control EVs. Relapse EVs were efficiently internalized by podocytes and induced significantly enhanced motility and albumin permeability. Moreover, relapse EVs induced significantly higher levels of RAC-GTP and phospho-p38 and decreased the levels of synaptopodin in podocytes. Circulating relapse EVs are biologically active molecules that carry active RAC1 as cargo and induce recapitulation of the NS phenotype in podocytes in vitro.NEW & NOTEWORTHY Up to now, the role of extracellular vesicles (EVs) in the pathogenesis of steroid-sensitive nephrotic syndrome (NS) has not been studied. Here, we found that relapse NS EVs contain significantly increased active RAC1, induce enhanced podocyte motility, and increase expression of RAC-GTP and phospho-p38 expression in vitro. These results suggest that plasma EVs are biologically active molecules in the pathogenesis of NS.


Subject(s)
Extracellular Vesicles/enzymology , Nephrotic Syndrome/enzymology , Podocytes/enzymology , rac1 GTP-Binding Protein/blood , Adolescent , Case-Control Studies , Cell Line , Child , Child, Preschool , Extracellular Vesicles/ultrastructure , Female , Humans , Male , Microfilament Proteins/metabolism , Nephrotic Syndrome/blood , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Phenotype , Phosphorylation , Podocytes/pathology , Recurrence , Remission Induction , Steroids/therapeutic use , Treatment Outcome , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Biosci Rep ; 41(7)2021 07 30.
Article in English | MEDLINE | ID: mdl-34240739

ABSTRACT

The serine protease prostasin is a negative regulator of lipopolysaccharide-induced inflammation and has a role in the regulation of cellular immunity. Prostasin expression in cancer cells inhibits migration and metastasis, and reduces epithelial-mesenchymal transition. Programmed death-ligand 1 (PD-L1) is a negative regulator of the immune response and its expression in cancer cells interferes with immune surveillance. The aim of the present study was to investigate if prostasin regulates PD-L1 expression. We established sublines overexpressing various forms of prostasin as well as a subline deficient for the prostasin gene from the Calu-3 human lung cancer cells. We report here that PD-L1 expression induced by interferon-γ (IFNγ) is further enhanced in cells overexpressing the wildtype membrane-anchored prostasin. The PD-L1 protein was localized on the cell surface and released into the culture medium in extracellular vesicles (EVs) with the protease-active prostasin. The epidermal growth factor-epidermal growth factor receptor (EGF-EGFR), protein kinase C (PKC), and mitogen-activated protein kinase (MAPK) participated in the prostasin-mediated up-regulation of PD-L1 expression. A Gene Set Enrichment Analysis (GSEA) of patient lung tumors in The Cancer Genome Atlas (TCGA) database revealed that prostasin and PD-L1 regulate common signaling pathways during tumorigenesis and tumor progression.


Subject(s)
Adenocarcinoma of Lung/enzymology , B7-H1 Antigen/metabolism , Extracellular Vesicles/enzymology , Lung Neoplasms/enzymology , Serine Endopeptidases/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , B7-H1 Antigen/genetics , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Vesicles/drug effects , Extracellular Vesicles/genetics , Extracellular Vesicles/immunology , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Serine Endopeptidases/genetics , Signal Transduction , Up-Regulation
10.
J Neurochem ; 157(6): 2195-2209, 2021 06.
Article in English | MEDLINE | ID: mdl-32880973

ABSTRACT

Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide, and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. HO-1 does not contain an N-terminal signal peptide and the mechanism responsible for its secretion remains unknown. Extracellular vesicles (EVs) are membrane-bound inclusions that transport microRNAs, messenger RNAs, lipids, and proteins among diverse cellular and extracellular compartments. The objective of the current study was to determine whether EVs in human biofluids contain HO-1, and whether the latter may be transported in EVs from brain to periphery. Total, L1 cell adhesion molecule protein (L1CAM)-enriched (neuron-derived), and glutamate aspartate transporter 1 (GLAST)-enriched (astrocyte-derived) EVs were purified from five different human biofluids (saliva [n = 40], plasma [n = 14], serum [n = 10], urine [n = 10], and cerebrospinal fluid [n = 11]) using polymer precipitation and immuno-affinity-based capture methods. L1CAM-enriched, GLAST-enriched, and L1CAM/GLAST-depleted (LGD) EV, along with EV-depleted (EVD), fractions were validated by nanoparticle tracking analysis, enzyme-linked immunosorbent assay (ELISA), and western blot. HO-1 was assayed in all fractions using ELISA and western blot. The majority of HO-1 protein was localized to LGD, L1CAM-enriched, and GLAST-enriched EVs of all human biofluids surveyed after adjusting for age and sex, with little HO-1 protein detected in EVD fractions. HO-1 protein in human biofluids is predominantly localized to EV compartments. A substantial proportion of EV HO-1 in peripheral human biofluids is derived from the central nervous system and may contribute to the systemic manifestations of various neurological conditions.


Subject(s)
Body Fluids/enzymology , Extracellular Vesicles/enzymology , Heme Oxygenase-1/metabolism , Adult , Aged , Biomarkers/metabolism , Body Fluids/chemistry , Extracellular Vesicles/chemistry , Female , Heme Oxygenase-1/analysis , Humans , Male , Middle Aged
12.
Biomed Pharmacother ; 133: 111048, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378955

ABSTRACT

Subarachnoid hemorrhage (SAH) is an acute and severe disease with high disability and mortality. Inflammatory reactions have been proven to occur throughout SAH. Extracellular vesicles derived from mesenchymal stem cells (MSCs-EVs) have shown broad potential for the treatment of brain dysfunction and neuroprotective effects through neurogenesis and angiogenesis after stroke. However, the mechanisms of EVs in neuroinflammation during the acute phase of SAH are not well known. Our present study was designed to investigate the effects of MSCs-EVs on neuroinflammation and the polarization regulation of microglia to the M2 phenotype and related signaling pathways after SAH in rats. The SAH model was induced by an improved method of intravascular perforation, and MSCs-EVs were injected via the tail vein. Post-SAH assessments included neurobehavioral tests as well as brain water content, immunohistochemistry, PCR and Western blot analyses. Our results showed that MSCs-EVs alleviated the expression of inflammatory cytokines in the parietal cortex and hippocampus 24 h and 48 h after SAH and that MSCs-EVs inhibited NF-κB and activated AMPK to reduce inflammation after SAH. Furthermore, MSC-EVs regulated the polarization of microglia toward the M2 phenotype by downregulating interleukin-1ß, cluster of differentiation 16, cluster of differentiation 11b, and inducible nitric oxide synthase and upregulating the expression of cluster of differentiation 206 and arginase-1. Additionally, MSCs-EVs inhibited the neuroinflammatory response and had neuroprotective effects in the brain tissues of rats after SAH. This study may support their use as a potential treatment strategy for early SAH in the future.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/enzymology , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Microglia/enzymology , NF-kappa B/metabolism , Subarachnoid Hemorrhage/surgery , Animals , Brain/pathology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Vesicles/enzymology , Male , Mesenchymal Stem Cells/enzymology , Microglia/pathology , Phenotype , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
13.
Vascul Pharmacol ; 135: 106807, 2020 12.
Article in English | MEDLINE | ID: mdl-33130246

ABSTRACT

AIM: Cell therapies are hampered by poor survival and growth of grafts. We tested whether forced co-expression of telomerase reverse transcriptase (TERT) and myocardin (MYOCD) improves post-infarct revascularization and tissue repair by adipose tissue-derived mesenchymal stromal cells (AT-MSCs). METHODS AND RESULTS: We transplanted AT-MSCs overexpressing MYOCD and TERT in a murine model of acute myocardial infarction (AMI). We characterized paracrine effects of AT-MSCs. When transplanted into infarcted hearts of C57BL/6 mice, AT-MSCs overexpressing TERT and MYOCD decreased scar tissue and the intra-scar CD3 and B220 lymphocyte infiltration; and increased arteriolar density as well as ejection fraction compared with saline or mock-transduced AT-MSCs. These effects were accompanied by higher persistence of the injected cells in the heart, increased numbers of Ki-67+ and CD117+ cells, and the expression of cardiac actin and ß-myosin heavy chain. Intramyocardial delivery of the secretome and its extracellular vesicle (EV)-enriched fraction also decreased scar tissue formation and increased arteriolar density in the murine AMI model. Proteomic analysis of AT-MSCs-EV-enriched fraction predicted the activation of vascular development and the inhibition of immune cell trafficking. Elevated concentrations of miR-320a, miR-150-5p and miR-126-3p associated with regulation of apoptosis and vasculogenesis were confirmed in the AT-MSCs-EV-enriched fraction. CONCLUSIONS: AT-MSCs overexpressing TERT and MYOCD promote persistence of transplanted aged AT-MSCs and enhance arteriolar density in a murine model of AMI. EV-enriched fraction is the component of the paracrine secretion by AT-MSCs with pro-angiogenic and anti-fibrotic activities.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/enzymology , Myocardial Infarction/surgery , Myocardium/metabolism , Nuclear Proteins/metabolism , Regeneration , Telomerase/metabolism , Trans-Activators/metabolism , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/enzymology , Extracellular Vesicles/transplantation , Fibrosis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Nuclear Proteins/genetics , Paracrine Communication , Recovery of Function , Signal Transduction , Telomerase/genetics , Trans-Activators/genetics
14.
Sci Rep ; 10(1): 19237, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159116

ABSTRACT

In the complex interplay of beneficial bacteria with the host, there are few examples of bacterial metabolites and effector molecules that have been consistently identified. Protective effects on the intestinal epithelium have been ascribed to P40 and P75, two well characterized cell wall muramidases, present in the culture supernatant of strains belonging to the taxon Lactobacillus casei/paracasei/rhamnosus. This work reports that Lactobacillus casei BL23 extracellular vesicles (BL23 EVs) have a small size (17-20 nm or 24-32 nm, depending on the method used) and contain lipoteichoic acid (LTA). Interestingly, all detected P40 and most of P75 were associated to EVs and possibly located at their external surface, as shown by proteinase K digestion. Biosensor assays showed that both proteins bind LTA and vesicles, suggesting that they could bind to ligands like LTA present on BL23 EVs. Native BL23 EVs have a moderate proinflammatory effect and they were able to induce phosphorylation of the epidermal growth factor receptor (EGFR), showing an effect similar to purified P40 and P75 and leading to the conclusion that the activity described in the supernatant (postbiotic) of these bacteria would be mainly due to P40 and P75 bound to EVs.


Subject(s)
Bacterial Proteins/pharmacology , Extracellular Vesicles/enzymology , Intestinal Mucosa/metabolism , Lacticaseibacillus casei/enzymology , Muramidase/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Humans
15.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32948029

ABSTRACT

The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.


Subject(s)
Antineoplastic Agents/therapeutic use , Chelating Agents/therapeutic use , Iron , Neoplasm Proteins/physiology , Peptide Hydrolases/physiology , Protease Inhibitors/therapeutic use , Zinc , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic , Chelating Agents/pharmacology , Disease Progression , Drug Design , Drug Screening Assays, Antitumor , Extracellular Fluid/enzymology , Extracellular Vesicles/enzymology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Kallikreins/antagonists & inhibitors , Kallikreins/physiology , Matrix Metalloproteinases/physiology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Oxaprozin/pharmacology , Oxaprozin/therapeutic use , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Phenylalanine/therapeutic use , Protease Inhibitors/pharmacology , Protein Kinases/physiology , Pyridines/pharmacology , Pyridines/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Thiosemicarbazones/pharmacology , Thiosemicarbazones/therapeutic use
16.
Mol Cancer Res ; 18(11): 1744-1754, 2020 11.
Article in English | MEDLINE | ID: mdl-32753475

ABSTRACT

The ATP6V1G1 subunit (V1G1) of the vacuolar proton ATPase (V-ATPase) pump is crucial for glioma stem cells (GSC) maintenance and in vivo tumorigenicity. Moreover, V-ATPase reprograms the tumor microenvironment through acidification and release of extracellular vesicles (EV). Therefore, we investigated the role of V1G1 in GSC small EVs and their effects on primary brain cultures. To this end, small EVs were isolated from patients-derived GSCs grown as neurospheres (NS) with high (V1G1HIGH-NS) or low (V1G1LOW-NS) V1G1 expression and analyzed for V-ATPase subunits presence, miRNA contents, and cellular responses in recipient cultures. Our results show that NS-derived small EVs stimulate proliferation and motility of recipient cells, with small EV derived from V1G1HIGH-NS showing the most pronounced activity. This involved activation of ERK1/2 signaling, in a response reversed by V-ATPase inhibition in NS-producing small EV. The miRNA profile of V1G1HIGH-NS-derived small EVs differed significantly from that of V1G1LOW-NS, which included miRNAs predicted to target MAPK/ERK signaling. Mechanistically, forced expression of a MAPK-targeting pool of miRNAs in recipient cells suppressed MAPK/ERK pathway activation and blunted the prooncogenic effects of V1G1HIGH small EV. These findings propose that the GSC influences the brain milieu through a V1G1-coordinated EVs release of MAPK/ERK-targeting miRNAs. Interfering with V-ATPase activity could prevent ERK-dependent oncogenic reprogramming of the microenvironment, potentially hampering local GBM infiltration. IMPLICATIONS: Our data identify a novel molecular mechanism of gliomagenesis specific of the GBM stem cell niche, which coordinates a V-ATPase-dependent reprogramming of the brain microenvironment through the release of specialized EVs.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MAP Kinase Signaling System , MicroRNAs/metabolism , Stem Cells/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Extracellular Vesicles/enzymology , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , MicroRNAs/genetics , Stem Cells/enzymology , Vacuolar Proton-Translocating ATPases/genetics
17.
Circ Res ; 127(7): 911-927, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32564697

ABSTRACT

RATIONALE: Vascular calcification, the formation of calcium phosphate crystals in the vessel wall, is mediated by vascular smooth muscle cells (VSMCs). However, the underlying molecular mechanisms remain elusive, precluding mechanism-based therapies. OBJECTIVE: Phenotypic switching denotes a loss of contractile proteins and an increase in migration and proliferation, whereby VSMCs are termed synthetic. We examined how VSMC phenotypic switching influences vascular calcification and the possible role of the uniquely calcium-dependent reactive oxygen species (ROS)-forming Nox5 (NADPH oxidase 5). METHODS AND RESULTS: In vitro cultures of synthetic VSMCs showed decreased expression of contractile markers CNN-1 (calponin 1), α-SMA (α-smooth muscle actin), and SM22-α (smooth muscle protein 22α) and an increase in synthetic marker S100A4 (S100 calcium binding protein A4) compared with contractile VSMCs. This was associated with increased calcification of synthetic cells in response to high extracellular Ca2+. Phenotypic switching was accompanied by increased levels of ROS and Ca2+-dependent Nox5 in synthetic VSMCs. Nox5 itself regulated VSMC phenotype as siRNA knockdown of Nox5 increased contractile marker expression and decreased calcification, while overexpression of Nox5 decreased contractile marker expression. ROS production in synthetic VSMCs was cytosolic Ca2+-dependent, in line with it being mediated by Nox5. Treatment of VSMCs with Ca2+ loaded extracellular vesicles (EVs) lead to an increase in cytosolic Ca2+. Inhibiting EV endocytosis with dynasore blocked the increase in cytosolic Ca2+ and VSMC calcification. Increased ROS production resulted in increased EV release and decreased phagocytosis by VSMCs. CONCLUSIONS: We show here that contractile VSMCs are resistant to calcification and identify Nox5 as a key regulator of VSMC phenotypic switching. Additionally, we describe a new mechanism of Ca2+ uptake via EVs and show that Ca2+ induces ROS production in VSMCs via Nox5. ROS production is required for release of EVs, which promote calcification. Identifying molecular pathways that control Nox5 and VSMC-derived EVs provides potential targets to modulate vascular remodeling and calcification in the context of mineral imbalance. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Cell Movement , Cell Proliferation , Extracellular Vesicles/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 5/metabolism , Reactive Oxygen Species/metabolism , Vascular Calcification/enzymology , Aged , Aged, 80 and over , Animals , Cells, Cultured , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 5/genetics , Phagocytosis , Phenotype , Signal Transduction , Sus scrofa , Vascular Calcification/genetics , Vascular Calcification/pathology
18.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326590

ABSTRACT

The identification of biomarkers for early diagnosis of Parkinson's disease (PD) is of pivotal importance for improving approaches for clinical intervention. The use of translatable animal models of pre-motor PD therefore offers optimal opportunities for novel biomarker discovery in vivo. Peptidylarginine deiminases (PADs) are a family of calcium-activated enzymes that contribute to protein misfolding through post-translational deimination of arginine to citrulline. Furthermore, PADs are an active regulator of extracellular vesicle (EV) release. Both protein deimination and extracellular vesicles (EVs) are gaining increased attention in relation to neurodegenerative diseases, including in PD, while roles in pre-motor PD have yet to be investigated. The current study aimed at identifying protein candidates of deimination in plasma and plasma-EVs in a rat model of pre-motor PD, to assess putative contributions of such post-translational changes in the early stages of disease. EV-cargo was further assessed for deiminated proteins as well as three key micro-RNAs known to contribute to inflammation and hypoxia (miR21, miR155, and miR210) and also associated with PD. Overall, there was a significant increase in circulating plasma EVs in the PD model compared with sham animals and inflammatory and hypoxia related microRNAs were significantly increased in plasma-EVs of the pre-motor PD model. A significantly higher number of protein candidates were deiminated in the pre-motor PD model plasma and plasma-EVs, compared with those in the sham animals. KEGG (Kyoto encyclopedia of genes and genomes) pathways identified for deiminated proteins in the pre-motor PD model were linked to "Alzheimer's disease", "PD", "Huntington's disease", "prion diseases", as well as for "oxidative phosphorylation", "thermogenesis", "metabolic pathways", "Staphylococcus aureus infection", gap junction, "platelet activation", "apelin signalling", "retrograde endocannabinoid signalling", "systemic lupus erythematosus", and "non-alcoholic fatty liver disease". Furthermore, PD brains showed significantly increased staining for total deiminated proteins in the brain vasculature in cortex and hippocampus, as well as increased immunodetection of deiminated histone H3 in dentate gyrus and cortex. Our findings identify EVs and post-translational protein deimination as novel biomarkers in early pre-motor stages of PD.


Subject(s)
Brain/metabolism , Citrullination , Extracellular Vesicles/metabolism , Parkinson Disease/blood , Protein-Arginine Deiminases/metabolism , Animals , Biomarkers/blood , Brain/physiopathology , Chromatography, Liquid , Disease Models, Animal , Extracellular Vesicles/enzymology , Extracellular Vesicles/ultrastructure , Immunohistochemistry , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Microscopy, Electron, Transmission , Parkinson Disease/enzymology , Parkinson Disease/metabolism , Protein Interaction Maps , Protein Processing, Post-Translational , Proteomics , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
19.
Ups J Med Sci ; 125(1): 10-18, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31774341

ABSTRACT

Background: Human seminal prostasomes are intrinsically heterogeneous extracellular vesicles (EVs) whose composition is, additionally, influenced by different physiological conditions. Aiming at the molecular properties of the prostasomal surface exemplified by glycan compositions as a possible distinction factor, we applied lectin-affinity chromatography (LAC) as a new tool for their separation. Since glycans, generally, exhibit various biological activities, introduction of glyco-parameters as reference could upgrade standardization of EVs isolated by different methods and intended for use in biomedicine.Methods: Preparations of seminal prostasomes from normozoospermic (sPro-N) and oligozoospermic (sPro-O) men were subjected to LAC on concanavalin A (Con A) and wheat germ agglutinin (WGA) columns. Prostasomes recovered in LAC-separated fractions were characterized according to the distribution of selected markers: gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), tetraspanin CD63, and total protein/glycoprotein composition.Results: Two CD63-immunoreactive populations exhibiting prostasome signature bands but differing in GGT activity and surface glycans were separated on the WGA column. Additional populations having distinct profiles of total glycoproteins and which can be tracked down by ALP activity were enriched on the Con A column. WGA-separated populations were similar in sPro-N and sPro-O, whereas Con A-separated ones were strikingly different.Conclusions: Membrane-associated gamma-glutamyl transferase and alkaline phosphatase in the context of Con A- and WGA-reactive glycans mark seminal prostasomes populations from normozoospermic and oligozoospermic men.


Subject(s)
Alkaline Phosphatase/metabolism , Concanavalin A/metabolism , Oligospermia/metabolism , Prostate/metabolism , Semen/metabolism , Wheat Germ Agglutinins/metabolism , gamma-Glutamyltransferase/metabolism , Case-Control Studies , Cell Membrane/enzymology , Chromatography, Affinity/methods , Extracellular Vesicles/enzymology , Extracellular Vesicles/metabolism , Humans , Male , Oligospermia/enzymology , Prostate/enzymology , Semen/enzymology
20.
Front Immunol ; 10: 1729, 2019.
Article in English | MEDLINE | ID: mdl-31404305

ABSTRACT

Extracellular adenine nucleotides participate in cell-to-cell communication and modulate the immune response. The concerted action of ectonucleotidases CD39 and CD73 plays a major role in the local production of anti-inflammatory adenosine, but both ectonucleotidases are rarely co-expressed by human T cells. The expression of CD39 on T cells increases upon T cell activation and is high at sites of inflammation. CD73, in contrast, disappears from the cellular membrane after activation. The possibility that CD73 could act in trans would resolve the conundrum of both enzymes being co-expressed for the degradation of ATP and the generation of adenosine. An enzymatically active soluble form of CD73 has been reported, and AMPase activity has been detected in body fluids of patients with inflammation and cancer. It is not yet clear how CD73, a glycosylphosphatidylinositol (GPI)-anchored protein, is released from the cell membrane, but plausible mechanisms include cleavage by metalloproteinases and shedding mediated by cell-associated phospholipases. Importantly, like many other GPI-anchored proteins, CD73 at the cell membrane is preferentially localized in detergent-resistant domains or lipid rafts, which often contribute to extracellular vesicles (EVs). Indeed, CD73-containing vesicles of different size and origin and with immunomodulatory function have been found in the tumor microenvironment. The occurrence of CD73 as non-cell-bound molecule widens the range of action of this enzyme at sites of inflammation. In this review, we will discuss the generation of non-cell-bound CD73 and its physiological role in inflammation.


Subject(s)
5'-Nucleotidase/physiology , Inflammation/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/genetics , Adenosine/physiology , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Cell Membrane/enzymology , Extracellular Fluid/metabolism , Extracellular Vesicles/enzymology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Glycosylphosphatidylinositols/metabolism , Humans , Inflammation/metabolism , Lymphocyte Activation , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Membrane Microdomains/enzymology , Mice , Neoplasm Proteins/physiology , Neoplasms/immunology , Neoplasms/pathology , Receptors, Purinergic P1/physiology , Solubility , Species Specificity , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...