Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Am J Respir Cell Mol Biol ; 70(1): 11-25, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37725486

ABSTRACT

The generation of bioactive truncated oxidized phospholipids (Tr-OxPLs) from oxidation of cell-membrane or circulating lipoproteins is a common feature of various pathological states. Scavenger receptor CD36 is involved in lipid transport and acts as a receptor for Tr-OxPLs. Interestingly, Tr-OxPLs and CD36 are involved in endothelial dysfunction-derived acute lung injury, but the precise mechanistic connections remain unexplored. In the present study, we investigated the role of CD36 in mediating pulmonary endothelial cell (EC) dysfunction caused by Tr-OxPLs. Our results demonstrated that the Tr-OxPLs KOdia-PC, Paz-PC, PGPC, PON-PC, POV-PC, and lysophosphocholine caused an acute EC barrier disruption as revealed by measurements of transendothelial electrical resistance and VE-cadherin immunostaining. More importantly, a synthetic amphipathic helical peptide, L37pA, targeting human CD36 strongly attenuated Tr-OxPL-induced EC permeability. L37pA also suppressed Tr-OxPL-induced endothelial inflammatory activation monitored by mRNA expression of inflammatory cytokines/chemokines and adhesion molecules. In addition, L37pA blocked Tr-OxPL-induced NF-κB activation and tyrosine phosphorylation of Src kinase and VE-cadherin. The Src inhibitor SU6656 attenuated KOdia-PC-induced EC permeability and inflammation, but inhibition of the Toll-like receptors (TLRs) TLR1, TLR2, TLR4, and TLR6 had no such protective effects. CD36-knockout mice were more resistant to Tr-OxPL-induced lung injury. Treatment with L37pA was equally effective in ameliorating Tr-OxPL-induced vascular leak and lung inflammation as determined by an Evans blue extravasation assay and total cell and protein content in BAL fluid. Altogether, these results demonstrate an essential role of CD36 in mediating Tr-OxPL-induced EC dysfunction and suggest a strong therapeutic potential of CD36 inhibitory peptides in mitigating lung injury and inflammation.


Subject(s)
Acute Lung Injury , Phospholipids , Animals , Mice , Humans , Phospholipids/metabolism , Acute Lung Injury/pathology , Inflammation , Peptides , Lung/pathology
2.
Cells ; 12(15)2023 07 26.
Article in English | MEDLINE | ID: mdl-37566016

ABSTRACT

Truncated phospholipid oxidation products (Tr-OxPL) increase in blood circulation with aging; however, their role in the severity of vascular dysfunction and bacterial lung injury in aging groups remains poorly understood. We investigated the effects of six Tr-OxPL species: KOdiA-PC, POVPC, PONPC, PGPC, Paz-PC, and Lyso-PC on endothelial dysfunction and lung inflammation caused by heat-killed Staphylococcus aureus (HKSA) in young (aged 2-4 months) and old (aged 12-18 months) mice, organotypic culture of precisely cut lung slices, and endothelial cells (mLEC) isolated from young and old mice. HKSA and Tr-OxPL combination caused a higher degree of vascular leak, the accumulation of inflammatory cells and protein in bronchoalveolar lavage, and inflammatory gene expression in old mice lungs. HKSA caused a greater magnitude of inflammatory gene activation in cell and ex vivo cultures from old mice, which was further augmented by Tr-OxPLs. L37pA peptide targeting CD36 receptor attenuated Tr-OxPL-induced endothelial cell permeability in young and old mLEC and ameliorated KOdiA-PC-induced vascular leak and lung inflammation in vivo. Finally, CD36 knockout mice showed better resistance to KOdiA-PC-induced lung injury in both age groups. These results demonstrate the aging-dependent vulnerability of pulmonary vasculature to elevated Tr-OxPL, which exacerbates bacterial lung injury. CD36 inhibition is a promising therapeutic approach for improving pneumonia outcomes in aging population.


Subject(s)
Lung Injury , Pneumonia , Animals , Mice , Phospholipids/metabolism , Endothelial Cells/metabolism , Lung Injury/metabolism , Pneumonia/metabolism , Aging
3.
Cell Signal ; 109: 110804, 2023 09.
Article in English | MEDLINE | ID: mdl-37437826

ABSTRACT

Oxidized phospholipids (OxPLs) are present at basal levels in circulation of healthy individuals, but a substantial increase and changes in composition of OxPLs may rapidly occur during microbial infections, sepsis, and trauma. Specifically, truncated oxidized phospholipids (Tr-OxPLs) exhibit detrimental effects on pulmonary endothelium, yet their role on modulation of lung injury caused by bacterial pathogens remains to be elucidated. This study investigated the effects of Tr-OxPL species: KOdiA-PC, POV-PC, PON-PC, PAz-PC, PGPC, and Lyso-PC on endothelial permeability and inflammatory responses to gram-positive bacterial particles. Results showed that all six tested Tr-OxPLs augmented endothelial barrier disruption caused by heat-killed Staphylococcus aureus (HKSA) as determined by VE-cadherin immunostaining and monitoring transendothelial electrical resistance. In parallel, even moderate elevation of Tr-OxPLs augmented HKSA-induced activation of NF-κB, secretion of IL-6 and IL-8, and protein expression of ICAM-1 and VCAM-1. In the mouse model of acute lung injury caused by intranasal injection of HKSA, intravenous Tr-OxPLs administration augmented HKSA-induced increase in BAL protein content and cell counts, tissue expression of TNFα, KC, IL1ß, and CCL2, and promoted vascular leak monitored by lung infiltration of Evans Blue. These results suggest that elevated Tr-OxPLs act as critical risk factor worsening bacterial pathogen-induced endothelial dysfunction and lung injury.


Subject(s)
Acute Lung Injury , Phospholipids , Animals , Mice , Phospholipids/metabolism , Phospholipids/pharmacology , Endothelium/metabolism , Lung/metabolism , Acute Lung Injury/metabolism , Intercellular Adhesion Molecule-1/metabolism , Oxidation-Reduction
4.
FASEB J ; 36(9): e22470, 2022 09.
Article in English | MEDLINE | ID: mdl-35969180

ABSTRACT

Extracellular DNA-binding proteins such as histones are danger-associated molecular pattern released by the injured tissues in trauma and sepsis settings, which trigger host immune response and vascular dysfunction. Molecular events leading to histone-induced endothelial cell (EC) dysfunction remain poorly understood. This study performed comparative analysis of H1, H2A, H2B, H3, and H4 histone subunits effects on human pulmonary EC permeability and inflammatory response. Analysis of transendothelial electrical resistance and EC monolayer permeability for macromolecues revealed that H3 and H4, but not H1, H2A, or H2B caused dose-dependent EC permeability accompanied by disassembly of adherens junctions. At higher doses, H3 and H4 activated nuclear factor kappa B inflammatory cascade leading to upregulation EC adhesion molecules ICAM1, VCAM1, E-selectin, and release of inflammatory cytokines. Inhibitory receptor analysis showed that toll-like receptor (TLR) 4 but not TLR1/2 or receptor for advanced glycation end inhibition significantly attenuated deleterious effects of H3 and H4 histones. Inhibitor of Rho-kinase was without effect, while inhibition of Src kinase caused partial preservation of cell-cell junctions, H3/H4-induced permeability and inflammation. Deleterious effects of H3/H4 were blocked by heparin. Activation of Epac-Rap1 signaling restored EC barrier properties after histone challenge. Intravenous injection of histones in mice caused elevation of inflammatory markers and increased vascular leak. Post-treatment with pharmacological Epac/Rap1 activator suppressed injurious effects of histones in vitro and in vivo. These results identify H3 and H4 as key histone subunits exhibiting deleterious effects on pulmonary vascular endothelium via TLR4-dependent mechanism. In conclusion, elevation of circulating histones may represent a serious risk of exacerbated acute lung injury (ALI) and multiple organ injury during severe trauma and infection.


Subject(s)
Histones , Inflammation , Animals , Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Histones/metabolism , Humans , Inflammation/metabolism , Mice , Permeability
5.
Am J Physiol Lung Cell Mol Physiol ; 323(3): L223-L239, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35852995

ABSTRACT

Extracellular histones released into the circulation following trauma, sepsis, and ARDS may act as potent damage-associated molecular pattern signals leading to multiple organ failure. Endothelial cell (EC) dysfunction caused by extracellular histones has been demonstrated in vitro and in vivo; however, precise mechanistic details of histone-induced EC dysfunction and exacerbation of ongoing inflammation remain poorly understood. This study investigated the role of extracellular histones in exacerbating preexisting endothelial dysfunction and acute lung injury. Histone subunits H3 and H4, but not H1, H2A, or H2B, induced permeability in human pulmonary EC. H3 and H4 at concentrations above 30 µg/mL caused EC inflammation reflected by activation of the NF-κB pathway, transcriptional activation, and release of cytokines and chemokines including IL-6 and IL-8, and increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1. Pharmacological inhibitors targeting Toll-like receptor TLR4 but not TLR2/6, blocked histone-induced EC dysfunction. H3 and H4 also strongly augmented EC permeability and inflammation caused by Gram-negative and Gram-positive bacterial particles, endotoxin, and TNFα. Heparin blocked histone-induced augmentation of EC inflammation caused by endotoxin and TNFα. Injection of histone in mouse models of lung injury caused by bacterial wall lipopolysaccharide (LPS) and heat-killed Staphylococcus aureus (HKSA) augmented ALI parameters: increased protein content, cell count, and inflammatory cytokine secretion in bronchoalveolar lavage fluid. Important clinical significance of these findings is in the demonstration that even a modest increase in extracellular histone levels can act as a severe exacerbating factor in conjunction with other EC barrier disruptive or proinflammatory agents.


Subject(s)
Acute Lung Injury , Histones , Acute Lung Injury/metabolism , Animals , Humans , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Am J Respir Cell Mol Biol ; 67(3): 375-388, 2022 09.
Article in English | MEDLINE | ID: mdl-35679261

ABSTRACT

TLR7 (Toll-like receptor 7), the sensor for single-stranded RNA, contributes to systemic inflammation and mortality in murine polymicrobial sepsis. Recent studies show that extracellular miR-146a-5p serves as a TLR7 ligand and plays an important role in regulating host innate immunity. However, the role of miR-146a-5p and TLR7 signaling in pulmonary inflammation, endothelial activation, and sepsis-associated acute respiratory distress syndrome remains unclear. Here, we show that intratracheal administration of exogenous miR-146a-5p in mice evokes lung inflammation, activates endothelium, and increases endothelial permeability via TLR7-dependent mechanisms. TLR7 deficiency attenuates pulmonary barrier dysfunction and reduces lung inflammatory response in a murine sepsis model. Moreover, the impact of miR-146a-5p-TLR7 signaling on endothelial activation appears to be a secondary effect because TLR7 is undetectable in the human pulmonary artery and microvascular endothelial cells (ECs), which show no response to direct miR-146a-5p treatment in vitro. Both conditioned media of miR-146a-5p-treated macrophages (Mϕ) and septic sera of wild-type mice induce a marked EC barrier disruption in vitro, whereas Mϕ conditioned media or septic sera of TLR7-/- mice do not exhibit such effect. Cytokine array and pathway enrichment analysis of the Mϕ conditioned media and septic sera identify TNFα (tumor necrosis factor α) as the main downstream effector of miR-146a-5p-TLR7 signaling responsible for the EC barrier dysfunction, which is further supported by neutralizing anti-TNFα antibody intervention. Together, these data demonstrate that TLR7 activation elicits pulmonary inflammation and endothelial barrier disruption by sensing extracellular miR-146a-5p and contributes to sepsis-associated acute respiratory distress syndrome.


Subject(s)
Membrane Glycoproteins , MicroRNAs , Respiratory Distress Syndrome , Sepsis , Toll-Like Receptor 7 , Animals , Culture Media, Conditioned , Endothelial Cells/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Respiratory Distress Syndrome/immunology , Sepsis/complications , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
7.
FASEB J ; 35(4): e21388, 2021 04.
Article in English | MEDLINE | ID: mdl-33724556

ABSTRACT

Suppressors of cytokine signaling (SOCS) provide negative regulation of inflammatory reaction. The role and precise cellular mechanisms of SOCS1 in control of endothelial dysfunction and barrier compromise associated with acute lung injury remain unexplored. Our results show that siRNA-mediated SOCS1 knockdown augmented lipopolysaccharide (LPS)-induced pulmonary endothelial cell (EC) permeability and enhanced inflammatory response. Consistent with in vitro data, EC-specific SOCS1 knockout mice developed more severe lung vascular leak and accumulation of inflammatory cells in bronchoalveolar lavage fluid. SOCS1 overexpression exhibited protective effects against LPS-induced endothelial permeability and inflammation, which were dependent on microtubule (MT) integrity. Biochemical and image analysis of unstimulated EC showed SOCS1 association with the MT, while challenge with LPS or MT depolymerizing agent colchicine impaired this association. SOCS1 directly interacted with N2 domains of MT-associated proteins CLIP-170 and CLASP2. Furthermore, N-terminal region of SOCS1 was indispensable for these interactions and SOCS1-ΔN mutant lacking N-terminal 59 amino acids failed to rescue LPS-induced endothelial dysfunction. Depletion of endogenous CLIP-170 or CLASP2 abolished SOCS1 interaction with Toll-like receptor-4 and Janus kinase-2 leading to impairment of SOCS1 inhibitory effects on LPS-induced inflammation. Altogether, these findings suggest that endothelial barrier protective and anti-inflammatory effects of SOCS1 are critically dependent on its targeting to the MT.


Subject(s)
Endothelial Cells/drug effects , Lipopolysaccharides/toxicity , Suppressor of Cytokine Signaling 1 Protein/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Cell Line , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Inflammation/chemically induced , Mice , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics
8.
J Biol Chem ; 296: 100239, 2021.
Article in English | MEDLINE | ID: mdl-33372035

ABSTRACT

Proinflammatory cytokines such as IL-6 induce endothelial cell (EC) barrier disruption and trigger an inflammatory response in part by activating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. The protein suppressor of cytokine signaling-3 (SOCS3) is a negative regulator of JAK-STAT, but its role in modulation of lung EC barrier dysfunction caused by bacterial pathogens has not been investigated. Using human lung ECs and EC-specific SOCS3 knockout mice, we tested the hypothesis that SOCS3 confers microtubule (MT)-mediated protection against endothelial dysfunction. SOCS3 knockdown in cultured ECs or EC-specific SOCS3 knockout in mice resulted in exacerbated lung injury characterized by increased permeability and inflammation in response to IL-6 or heat-killed Staphylococcus aureus (HKSA). Ectopic expression of SOCS3 attenuated HKSA-induced EC dysfunction, and this effect required assembled MTs. SOCS3 was enriched in the MT fractions, and treatment with HKSA disrupted SOCS3-MT association. We discovered that-in addition to its known partners gp130 and JAK2-SOCS3 interacts with MT plus-end binding proteins CLIP-170 and CLASP2 via its N-terminal domain. The resulting SOCS3-CLIP-170/CLASP2 complex was essential for maximal SOCS3 anti-inflammatory effects. Both IL-6 and HKSA promoted MT disassembly and disrupted SOCS3 interaction with CLIP-170 and CLASP2. Moreover, knockdown of CLIP-170 or CLASP2 impaired SOCS3-JAK2 interaction and abolished the anti-inflammatory effects of SOCS3. Together, these findings demonstrate for the first time an interaction between SOCS3 and CLIP-170/CLASP2 and reveal that this interaction is essential to the protective effects of SOCS3 in lung endothelium.


Subject(s)
Inflammation/genetics , Lung Injury/genetics , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Acute Lung Injury/genetics , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Cytoskeleton/genetics , Endothelial Cells , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology , Endothelium, Vascular/pathology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intercellular Junctions/genetics , Interleukin-6/genetics , Lung Injury/metabolism , Lung Injury/microbiology , Lung Injury/pathology , Mice , Mice, Knockout , Permeability , Staphylococcus aureus/pathogenicity
9.
Sci Rep ; 10(1): 12497, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32719338

ABSTRACT

Acute lung injury (ALI), a common condition in critically ill patients, has limited treatments and high mortality. Aging is a risk factor for ALI. Sirtuins (SIRTs), central regulators of the aging process, decrease during normal aging and in aging-related diseases. We recently showed decreased SIRT7 expression in lung tissues and fibroblasts from patients with pulmonary fibrosis compared to controls. To gain insight into aging-related mechanisms in ALI, we investigated the effects of SIRT7 depletion on lipopolysaccharide (LPS)-induced inflammatory responses and endothelial barrier permeability in human primary pulmonary endothelial cells. Silencing SIRT7 in pulmonary artery or microvascular endothelial cells attenuated LPS-induced increases in ICAM1, VCAM1, IL8, and IL6 and induced endomesenchymal transition (EndoMT) with decreases in VE-Cadherin and PECAM1 and increases in collagen, alpha-smooth muscle actin, TGFß receptor 1, and the transcription factor Snail. Loss of endothelial adhesion molecules was accompanied by increased F-actin stress fibers and increased endothelial barrier permeability. Together, these results show that an aging phenotype induced by SIRT7 deficiency promotes EndoMT with impaired inflammatory responses and dysfunction of the lung vascular barrier.


Subject(s)
Capillary Permeability , Endothelial Cells/pathology , Epithelium/pathology , Inflammation/metabolism , Lung/pathology , Sirtuins/deficiency , Adult , Animals , Bleomycin , Cell Membrane Permeability , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Sirtuins/genetics , Sirtuins/metabolism , Transforming Growth Factor beta/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 318(3): L533-L548, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913681

ABSTRACT

Transfusion of red blood cells (RBCs) is a common life-saving clinical practice in severely anemic or hemorrhagic patients; however, it may result in serious pathological complications such as transfusion-related acute lung injury. The factors mediating the deleterious effects of RBC transfusion remain unclear. In this study, we tested the effects of washed long-term (RBC-O; >28 days) versus short-term (RBC-F; <14 days) stored RBCs and their supernatants on lung endothelial (EC) permeability under control and inflammatory conditions. RBCs enhanced basal EC barrier function as evidenced by an increase in transendothelial electrical resistance and decrease in permeability for macromolecules. RBCs also attenuated EC hyperpermeability and suppressed secretion of EC adhesion molecule ICAM-1 and proinflammatory cytokine IL-8 in response to LPS or TNF-α. In both settings, RBC-F had slightly higher barrier protective effects as compared with RBC-O. In contrast, supernatants from both RBC-F and RBC-O disrupted the EC barrier. The early phase of EC permeability response caused by RBC supernatants was partially suppressed by antioxidant N-acetyl cysteine and inhibitor of Src kinase family PP2, while addition of heme blocker and inhibition of NOD-like receptor family pyrin domain containing protein 3 (NLRP3), stress MAP kinases, receptor for advanced glycation end-products (RAGE), or Toll-like receptor-4 (TLR4) signaling were without effect. Morphological analysis revealed that RBC supernatants increased LPS- and TNF-α-induced breakdown of intercellular junctions and formation of paracellular gaps. RBC supernatants augmented LPS- and TNF-α-induced EC inflammation reflected by increased production of IL-6, IL-8, and soluble ICAM-1. These findings demonstrate the deleterious effects of RBC supernatants on EC function, which may have a major impact in pathological consequences associated with RBC transfusion.


Subject(s)
Blood Preservation/adverse effects , Cell Membrane Permeability , Endothelium, Vascular/pathology , Erythrocytes/pathology , Inflammation/pathology , Lung/pathology , Allogeneic Cells , Blood Component Removal/methods , Endothelium, Vascular/immunology , Erythrocyte Transfusion/adverse effects , Humans , Inflammation/etiology , Inflammation/immunology , Lung/immunology
11.
Cytokine ; 110: 110-115, 2018 10.
Article in English | MEDLINE | ID: mdl-29729649

ABSTRACT

Interstitial cystitis (IC) is a chronic syndrome that affects the urinary bladder. The etiology of this disease is unclear, and no effective therapies are available at this time. Although inflammation is suspected, no clear evidence for a role of conventional mediators of inflammation, such as cytokines and their downstream molecules, has been obtained to date. Our previous studies indicated that primary cell cultures derived from IC urothelium abnormally express molecules associated with cell adhesion. Here we describe a mechanism by which transcriptional changes in tight junction and adhesion molecules are mediated. Oncosuppressor proteins p53 and cyclin-dependent protein kinase inhibitor p21 directly associate with regulatory sites on the ZO-1 and E-cadherin genes, identifying important roles for p53 and p21 in driving non-oncogenic pathologies. These data also suggest that interference with these factors offers a potential therapeutic opportunity.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cystitis, Interstitial/metabolism , Gene Expression/physiology , Tumor Suppressor Protein p53/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Line , Cytokines/metabolism , Humans , Inflammation/metabolism , Tight Junctions/metabolism , Tight Junctions/physiology , Transcription, Genetic/physiology , Urinary Bladder/metabolism , Urinary Bladder/physiology , Urothelium/metabolism , Zonula Occludens-1 Protein/metabolism
12.
ACS Omega ; 2(9): 5618-5632, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28983523

ABSTRACT

Glycoamino acid analogues of the Thomsen-Friedenreich antigen disaccharide, where the 4' and 4″ hydroxyl groups were substituted with fluorine or hydrogen, were synthesized and incorporated into the asialylated antiproliferative factor (as-APF), a biologically active form of APF, a glycopeptide found in the urine of patients with interstitial cystitis. Various strategies were employed to incorporate the fluorine atom at the 4-positions of either the galactose or N-acetylgalactosamine unit of the disaccharide antigen, based on stereochemistry and reactivity. These glycopeptides were evaluated in antiproliferative assays on both primary normal bladder epithelial cells and T24 bladder carcinoma cells. Unlike many previously published substitutions to APF, mono-4'-fluorination of the GalNAc residue did not affect the activity, whereas fluoro-derivatives of the galactose 4″-position or both 4' and 4″ hydroxyls showed a reduced potency relative to the monosubstituted GalNAc derivative. A fourth compound where the 4″ position of galactose was deoxygenated showed a lower potency than the parent and monosubstituted compounds. These results suggest that specific substitutions in the sugar moieties in the APF can be tolerated, and the glycomimetic design of APF analogues can include fluorine in the GalNAc sugar of the disaccharide.

13.
BJU Int ; 118(1): 161-72, 2016 07.
Article in English | MEDLINE | ID: mdl-26919663

ABSTRACT

OBJECTIVES: To determine whether protein kinase B (Akt) signalling and secretion of specific downstream effector proteins are abnormal in specific cell fractions of bladder epithelial cells from patients with interstitial cystitis/bladder pain syndrome (IC/BPS), as explanted bladder epithelial cells from patients with IC/BPS produce a frizzled 8-related glycopeptide antiproliferative factor (APF) that inhibits normal bladder epithelial cell proliferation and expression of several proteins known to be regulated by Akt signalling. A related secondary objective was to determine whether treatment of normal bladder epithelial cells with active synthetic asialo-antiproliferative factor (as-APF) induces similar changes in Akt signalling and specific downstream effector proteins/mRNAs. PATIENTS AND METHODS: Cell proteins were extracted into four subcellular fractions from primary bladder epithelial explants of six patients who fulfilled modified National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) criteria for IC/BPS and six age- and gender-matched controls. Total and/or phosphorylated cellular Akt, glycogen synthase kinase 3ß (GSK3ß), and ß-catenin; total cellular JunB; and secreted matrix metalloproteinase 2 (MMP2) and heparin-binding epidermal growth factor-like growth factor (HB-EGF) levels were determined by Western blot. MMP2, JunB, p53, uroplakin 3 (UPK3), and ß-actin mRNAs were quantified by quantitative reverse transcriptase-polymerase chain reaction. Akt activity was determined by nonradioactive assay. RESULTS: IC/BPS cells had lower Akt activity, along with lower Akt ser473- and GSK3ß ser9-phosphorylation and higher ß-catenin ser33,37/thr41-phosphorylation in specific fractions as compared with matched control cells. IC/BPS explants also had evidence of additional downstream abnormalities compared with control cells, including lower nuclear JunB; lower secreted MMP2 and HB-EGF; plus lower MMP2, JunB, and UPK3 mRNAs but higher p53 mRNA relative to ß-actin. Each of these IC/BPS cell abnormalities was also induced in normal cells by as-APF. CONCLUSION: These findings indicate that IC/BPS cells have abnormal Akt activity with downstream protein expression abnormalities including decreased MMP2 and HB-EGF secretion. They also support the hypothesis that APF plays a role in the pathogenesis of IC/BPS via its effects on cell Akt signalling and HB-EGF production.


Subject(s)
Cystitis, Interstitial/physiopathology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Urinary Bladder/physiopathology , Urothelium/physiopathology , Adult , Cells, Cultured , Cystitis, Interstitial/pathology , Female , Humans , Middle Aged , Urinary Bladder/pathology , Urothelium/pathology
14.
Invest New Drugs ; 30(5): 1849-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21931970

ABSTRACT

Antiproliferative factor (APF) is a potent frizzled protein 8-related sialoglycopeptide inhibitor of bladder epithelial cell proliferation that mediates its activity by binding to cytoskeletal associated protein 4 in the cell membrane. Synthetic asialylated APF (as-APF) (Galß1-3GalNAcα-O-TVPAAVVVA) was previously shown to inhibit both normal bladder epithelial as well as T24 bladder carcinoma cell proliferation and heparin-binding epidermal growth factor-like growth factor (HB-EGF) production at low nanomolar concentrations, and an L: -pipecolic acid derivative (Galß1-3GalNAcα-O-TV-pipecolic acid-AAVVVA) was also shown to inhibit normal bladder epithelial cell proliferation. To better determine their spectrum of activity, we measured the effects of these APF derivatives on the proliferation of cells derived from additional urologic carcinomas (bladder and kidney), non-urologic carcinomas (ovary, lung, colon, pancreas, and breast), and melanomas using a (3)H-thymidine incorporation assay. We also measured the effects of as-APF on cell HB-EGF and matrix metalloproteinase (MMP2) secretion plus cell invasion, using qRT-PCR, Western blot and an in vitro invasion assay. L: -pipecolic acid as-APF and/or as-APF significantly inhibited proliferation of each cell line in a dose-dependent manner with IC(50)'s in the nanomolar range, regardless of tissue origin, cell type (carcinoma vs. melanoma), or p53 or ras mutation status. as-APF also inhibited HB-EGF and MMP2 production plus in vitro invasion of tested bladder, kidney, breast, lung, and melanoma tumor cell lines, in a dose-dependent manner (IC(50) = 1-100 nM). Synthetic APF derivatives are potent inhibitors of urologic and non-urologic carcinoma plus melanoma cell proliferation, MMP2 production, and invasion, and may be useful for development as adjunctive antitumor therapy(ies).


Subject(s)
Carcinoma/drug therapy , Glycoproteins/pharmacology , Melanoma/drug therapy , Receptors, Cell Surface/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Melanoma/metabolism , Melanoma/pathology , Neoplasm Invasiveness , Prognosis , Sialoglycoproteins/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology
15.
Chem Biol Drug Des ; 77(6): 421-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21352500

ABSTRACT

Interstitial cystitis/painful bladder syndrome is a chronic bladder disorder with epithelial thinning or ulceration, pain, urinary frequency and urgency, for which there is no reliably effective therapy. We previously reported that interstitial cystitis/painful bladder syndrome bladder epithelial cells make a glycopeptide antiproliferative factor or 'APF' (Neu5Acα2-3Galß1-3GalNAcα-O-TVPAAVVVA) that induces abnormalities in normal cells similar to those in interstitial cystitis/painful bladder syndrome cells in vitro, including decreased proliferation, decreased tight junction formation, and increased paracellular permeability. We screened inactive APF derivatives for their ability to block antiproliferative activity of asialylated-APF ('as-APF') in normal bladder cells and determined the ability of as-APF-blocking derivatives to normalize tight junction protein expression, paracellular permeability, and/or proliferation of interstitial cystitis/painful bladder syndrome cells. Only two of these derivatives [Galß1-3GalNAcα-O-TV-(d-pipecolic acid)-AAVVVA and Galß1-3GalNAcα-O-TV-(d-proline)-AAVVVA] blocked as-APF antiproliferative activity in normal cells (p < 0.001 for both). Both of these antagonists also 1) significantly increased mRNA expression of ZO-1, occludin, and claudins 1, 4, 8, and 12 in interstitial cystitis/painful bladder syndrome cells by qRT-PCR; 2) normalized interstitial cystitis/painful bladder syndrome epithelial cell tight junction protein expression and tight junction formation by confocal immunofluorescence microscopy; and 3) decreased paracellular permeability of (14) C-mannitol and (3) H-inulin between confluent interstitial cystitis/painful bladder syndrome epithelial cells on Transwell plates, suggesting that these potent APF antagonists may be useful for the development as interstitial cystitis/painful bladder syndrome therapies.


Subject(s)
Cystitis, Interstitial/drug therapy , Epithelial Cells/drug effects , Glycoproteins/chemistry , Glycoproteins/pharmacology , Tight Junctions/drug effects , Urinary Bladder/cytology , Amino Acid Sequence , Carbohydrate Sequence , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cystitis, Interstitial/metabolism , Cystitis, Interstitial/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Pipecolic Acids/chemistry , Proline/chemistry , Tight Junctions/metabolism , Tight Junctions/pathology
16.
Mol Cell Proteomics ; 10(6): M110.007492, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21422242

ABSTRACT

Antiproliferative factor (APF), a Frizzled-8 protein-related sialoglycopeptide involved in the pathogenesis of interstitial cystitis, potently inhibits proliferation of normal urothelial cells as well as certain cancer cells. To elucidate the molecular mechanisms of the growth-inhibitory effect of APF, we performed stable isotope labeling by amino acids in cell culture analysis of T24 bladder cancer cells treated with and without APF. Among over 2000 proteins identified, 54 were significantly up-regulated and 48 were down-regulated by APF treatment. Bioinformatic analysis revealed that a protein network involved in cell adhesion was substantially altered by APF and that ß-catenin was a prominent node in this network. Functional assays demonstrated that APF down-regulated ß-catenin, at least in part, via proteasomal and lysosomal degradation. Moreover, silencing of ß-catenin mimicked the antiproliferative effect of APF whereas ectopic expression of nondegradable ß-catenin rescued growth inhibition in response to APF, confirming that ß-catenin is a key mediator of APF signaling. Notably, the key role of ß-catenin in APF signaling is not restricted to T24 cells, but was also observed in an hTERT-immortalized human bladder epithelial cell line, TRT-HU1. In addition, the network model suggested that ß-catenin is linked to cyclooxygenase-2 (COX-2), implying a potential connection between APF and inflammation. Functional assays verified that APF increased the production of prostaglandin E(2) and that down-modulation of ß-catenin elevated COX-2 expression, whereas forced expression of nondegradable ß-catenin inhibited APF-induced up-regulation of COX-2. Furthermore, we confirmed that ß-catenin was down-regulated whereas COX-2 was up-regulated in epithelial cells explanted from IC bladder biopsies compared with control tissues. In summary, our quantitative proteomics study describes the first provisional APF-regulated protein network, within which ß-catenin is a key node, and provides new insight that targeting the ß-catenin signaling pathway may be a rational approach toward treating interstitial cystitis.


Subject(s)
Glycoproteins/pharmacology , Inflammation Mediators/physiology , beta Catenin/metabolism , Cell Adhesion Molecules/metabolism , Cell Culture Techniques , Cell Line , Cell Proliferation , Cyclooxygenase 2/metabolism , Cystitis, Interstitial/metabolism , Down-Regulation , Humans , Inflammation Mediators/pharmacology , Intercellular Signaling Peptides and Proteins , Isotope Labeling , Metabolic Networks and Pathways , Proteomics , RNA Interference , Signal Transduction , Urinary Bladder/metabolism , Urinary Bladder/pathology , beta Catenin/genetics
17.
J Exp Clin Cancer Res ; 29: 160, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21143984

ABSTRACT

BACKGROUND: Urinary bladder cancer is a common malignancy worldwide, and outcomes for patients with advanced bladder cancer remain poor. Antiproliferative factor (APF) is a potent glycopeptide inhibitor of epithelial cell proliferation that was discovered in the urine of patients with interstitial cystitis, a disorder with bladder epithelial thinning and ulceration. APF mediates its antiproliferative activity in primary normal bladder epithelial cells via cytoskeletal associated protein 4 (CKAP4). Because synthetic asialo-APF (as-APF) has also been shown to inhibit T24 bladder cancer cell proliferation at nanomolar concentrations in vitro, and because the peptide segment of APF is 100% homologous to part of frizzled 8, we determined whether CKAP4 mediates as-APF inhibition of proliferation and/or downstream Wnt/frizzled signaling events in T24 cells. METHODS: T24 cells were transfected with double-stranded siRNAs against CKAP4 and treated with synthetic as-APF or inactive control peptide; cells that did not undergo electroporation and cells transfected with non-target (scrambled) double-stranded siRNA served as negative controls. Cell proliferation was determined by 3H-thymidine incorporation. Expression of Akt, glycogen synthase kinase 3ß (GSK3ß), ß-catenin, p53, and matrix metalloproteinase 2 (MMP2) mRNA was determined by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Akt, GSK-3ß, MMP2, ß-catenin, and p53 protein expression, plus Akt, GSK-3ß, and ß-catenin phosphorylation, were determined by Western blot. RESULTS: T24 cell proliferation, MMP2 expression, Akt ser473 and thr308 phosphorylation, GSK3ß tyr216 phosphorylation, and ß-catenin ser45/thr41 phosphorylation were all decreased by APF, whereas p53 expression, and ß-catenin ser33,37/thr41 phosphorylation, were increased by APF treatment in non-electroporated and non-target siRNA-transfected cells. Neither mRNA nor total protein expression of Akt, GSK3ß, or ß-catenin changed in response to APF in these cells. In addition, the changes in cell proliferation, MMP2/p53 mRNA and protein expression, and Akt/GSK3ß/ß-catenin phosphorylation in response to APF treatment were all specifically abrogated following CKAP4 siRNA knockdown. CONCLUSIONS: Synthetic as-APF inhibits cell proliferation in T24 bladder carcinoma cells via the CKAP4 receptor. The mechanism for this inhibition involves regulating phosphorylation of specific cell signaling molecules (Akt, GSK3ß, and ß-catenin) plus mRNA and protein expression of p53 and MMP2.


Subject(s)
Carcinoma, Transitional Cell/metabolism , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/metabolism , Blotting, Western , Carcinoma, Transitional Cell/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Glycoproteins/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
18.
ACS Med Chem Lett ; 1(8): 390-4, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-24900223

ABSTRACT

Antiproliferative factor (APF), a sialylated glycopeptide secreted by explanted bladder epithelial cells from interstitial cystitis/painful bladder syndrome (IC/PBS) patients, and its unsialylated analogue (as-APF) significantly decrease proliferation of bladder epithelial cells and/or certain carcinoma cell lines in vitro. We recently reported a structure-activity relationship profile for the peptide portion of as-APF and revealed that truncation of the C-terminal alanine did not significantly affect antiproliferative activity. To better understand the structural basis for the maintenance of activity of this truncated eight amino acid as-APF (as-APF8), we synthesized several amino acid-substituted derivatives and studied their ability to inhibit bladder epithelial cell proliferation in vitro as well as their solution conformations by CD and NMR spectroscopy. While single amino acid changes to as-APF8 often strongly reduced activity, full potency was retained when the trivaline tail was replaced with three alanines. The Ala(6-8) derivative 9 is the simplest, fully potent APF analogue synthesized to date.

19.
Mol Biol Cell ; 20(5): 1454-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19144824

ABSTRACT

Previously, we identified cytoskeleton-associated protein 4 (CKAP4) as a major substrate of the palmitoyl acyltransferase, DHHC2, using a novel proteomic method called palmitoyl-cysteine identification, capture and analysis (PICA). CKAP4 is a reversibly palmitoylated and phosphorylated protein that links the ER to the cytoskeleton. It is also a high-affinity receptor for antiproliferative factor (APF), a small sialoglycopeptide secreted from bladder epithelial cells of patients with interstitial cystitis (IC). The role of DHHC2-mediated palmitoylation of CKAP4 in the antiproliferative response of HeLa and normal bladder epithelial cells to APF was investigated. Our data show that siRNA-mediated knockdown of DHHC2 and consequent suppression of CKAP4 palmitoylation inhibited the ability of APF to regulate cellular proliferation and blocked APF-induced changes in the expression of E-cadherin, vimentin, and ZO-1 (genes known to play a role in cellular proliferation and tumorigenesis). Immunocytochemistry revealed that CKAP4 palmitoylation by DHHC2 is required for its trafficking from the ER to the plasma membrane and for its nuclear localization. These data suggest an important role for DHHC2-mediated palmitoylation of CKAP4 in IC and in opposing cancer-related cellular behaviors and support the idea that DHHC2 is a tumor suppressor.


Subject(s)
Acyltransferases/physiology , Glycoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Half-Life , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Lipoylation , Models, Biological , Protein Transport , RNA Interference , RNA, Messenger/metabolism , Tumor Suppressor Proteins/metabolism
20.
J Med Chem ; 51(19): 5974-83, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18788730

ABSTRACT

We performed comprehensive structure-activity relationship (SAR) studies on the peptide portion of antiproliferative factor (APF), a sialylated frizzled-8 related glycopeptide that inhibits normal bladder epithelial and urothelial carcinoma cell proliferation. Glycopeptide derivatives were synthesized by solid-phase methods using standard Fmoc chemistry and purified by RP-HPLC; all intermediate and final products were verified by HPLC-MS and NMR analyses. Antiproliferative activity of each derivative was determined by inhibition of (3)H-thymidine incorporation in primary normal human bladder epithelial cells. Structural components of the peptide segment of APF that proved to be important for biological activity included the presence of at least eight of the nine N-terminal amino acids, a negative charge in the C-terminal amino acid, a free amino group at the N-terminus, maintenance of a specific amino acid sequence in the C-terminal tail, and trans conformation for the peptide bonds. These data provide critical guidelines for optimization of structure in design of APF analogues as potential therapeutic agents.


Subject(s)
Cystitis, Interstitial/urine , Epithelial Cells/drug effects , Glycopeptides/pharmacology , Glycoproteins/pharmacology , Growth Inhibitors/pharmacology , Urinary Bladder/cytology , Adolescent , Amino Acid Sequence , Amino Acids/chemistry , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Epithelial Cells/chemistry , Glycopeptides/chemical synthesis , Glycopeptides/chemistry , Glycoproteins/chemical synthesis , Glycoproteins/chemistry , Growth Inhibitors/chemical synthesis , Growth Inhibitors/chemistry , Humans , Intercellular Signaling Peptides and Proteins , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship , Urinary Bladder/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...