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










Publication year range
1.
Int J Mol Sci ; 25(9)2024 May 05.
Article in English | MEDLINE | ID: mdl-38732252

ABSTRACT

Several studies have shown an inverse correlation between the likelihood of developing a neurodegenerative disorder and cancer. We previously reported that the levels of amyloid beta (Aß), at the center of Alzheimer's disease pathophysiology, are regulated by acetylcholinesterase (AChE) in non-small cell lung cancer (NSCLC). Here, we examined the effect of Aß or its fragments on the levels of ACh in A549 (p53 wild-type) and H1299 (p53-null) NSCLC cell media. ACh levels were reduced by cell treatment with Aß 1-42, Aß 1-40, Aß 1-28, and Aß 25-35. AChE and p53 activities increased upon A549 cell treatment with Aß, while knockdown of p53 in A549 cells increased ACh levels, decreased AChE activity, and diminished the Aß effects. Aß increased the ratio of phospho/total p38 MAPK and decreased the activity of PKC. Inhibiting p38 MAPK reduced the activity of p53 in A549 cells and increased ACh levels in the media of both cell lines, while opposite effects were found upon inhibiting PKC. ACh decreased the activity of p53 in A549 cells, decreased p38 MAPK activity, increased PKC activity, and diminished the effect of Aß on those activities. Moreover, the negative effect of Aß on cell viability was diminished by cell co-treatment with ACh.


Subject(s)
Acetylcholine , Amyloid beta-Peptides , Carcinoma, Non-Small-Cell Lung , Cell Survival , Lung Neoplasms , Protein Kinase C , Tumor Suppressor Protein p53 , p38 Mitogen-Activated Protein Kinases , Humans , A549 Cells , Acetylcholine/metabolism , Acetylcholine/pharmacology , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Survival/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Sci Rep ; 14(1): 4921, 2024 02 28.
Article in English | MEDLINE | ID: mdl-38418632

ABSTRACT

Previously, we found that the levels of soluble amyloid precursor protein α (sAPPα) are regulated, in part, by acetylcholinesterase (AChE) in human A549 (p53 wild-type) and H1299 (p53-null) NSCLC cell lines. In this study, we found regulation of sAPPα levels in the media by leptin, a widely recognized obesity-associated adipokine that has recently been shown to play a possible role in cancer signaling. Increased levels of sAPPα, that were accompanied by lower Aß40/42 levels in the media of A549 and H1299 cells, were detected upon cell incubation with leptin. Conversely, knockdown of leptin or its receptor led to reduced levels of sAPPα and increased levels of Aß40/42 in the media of A549 and H1299 cells, suggesting that leptin likely shifts APP processing toward the non-amyloidogenic pathway. A549 cell treatment with leptin increased acetylcholine levels and blocked the activities of AChE and p53. Treatment with leptin resulted in increased activation of PKC, ERK1/2, PI3K, and the levels of sAPPα, effects that were reversed by treatment with kinase inhibitors and/or upon addition of AChE to A549 and H1299 cell media. Cell viability increased by treatment of A549 and H1299 cells with leptin and decreased upon co-treatment with AChE and/or inhibitors targeting PKC, ERK1/2, and PI3K. This study is significant as it provides evidence for a likely carcinogenic role of leptin in NSCLC cells via upregulation of sAPPα levels in the media, and highlights the importance of targeting leptin as a potential therapeutic strategy for NSCLC treatment.


Subject(s)
Amyloid beta-Protein Precursor , Lung Neoplasms , Humans , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Leptin/metabolism , Acetylcholinesterase/metabolism , Tumor Suppressor Protein p53/genetics , Phosphatidylinositol 3-Kinases/metabolism
3.
Cancers (Basel) ; 15(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38067204

ABSTRACT

In addition to binding to nicotinic acetylcholine receptors (nAChRs), nicotine is known to regulate the ß-adrenergic receptors (ß-ARs) promoting oncogenic signaling. Using A549 (p53 wild-type) and H1299 (p53-null) lung cancer cells, we show that nicotine treatment led to: increased adrenaline/noradrenaline levels, an effect blocked by treatment with the α7nAChR inhibitor (α-BTX) but not by the ß-blocker (propranolol) or the α4ß2nAChR antagonist (DhßE); decreased GABA levels in A549 and H1299 cell media, an effect blocked by treatment with DhßE; increased VEGF levels and PI3K/AKT activities, an effect diminished by cell co-treatment with α-BTX, propranolol, and/or DhßE; and inhibited p53 activity in A549 cells, that was reversed, upon cell co-treatment with α-BTX, propranolol, and/or DhßE or by VEGF immunodepletion. VEGF levels increased upon cell treatment with nicotine, adrenaline/noradrenaline, and decreased with GABA treatment. On the other hand, the p53 activity decreased in A549 cells treated with nicotine, adrenaline/noradrenaline and increased upon cell incubation with GABA. Knockdown of p53 led to increased VEGF levels in the media of A549 cells. The addition of anti-VEGF antibodies to A549 and H1299 cells decreased cell viability and increased apoptosis; blocked the activities of PI3K, AKT, and NFκB in the absence or presence of nicotine; and resulted in increased p53 activation in A549 cells. We conclude that VEGF can be upregulated via α7nAChR and/or ß-ARs and downregulated via GABA and/or p53 in response to the nicotine treatment of NSCLC cells.

4.
Biomedicines ; 11(9)2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37760996

ABSTRACT

The ectodomain of the transmembrane protein E-cadherin can be cleaved and released in a soluble form referred to as soluble E-cadherin, or sE-cad, accounting for decreased E-cadherin levels at the cell surface. Among the proteases implicated in this cleavage are matrix metalloproteases (MMP), including MMP9. Opposite functions have been reported for full-length E-cadherin and sE-cad. In this study, we found increased MMP9 levels in the media of two non-small cell lung cancer (NSCLC) cell lines, A549 and H1299, treated with BDNF, nicotine, or epinephrine that were decreased upon cell treatment with the ß-adrenergic receptor blocker propranolol. Increased MMP9 levels correlated with increased sE-cad levels in A549 cell media, and knockdown of MMP9 in A549 cells led to downregulation of sE-cad levels in the media. Previously, we reported that A549 and H1299 cell viability increased with nicotine and/or BDNF treatment and decreased upon treatment with propranolol. In investigating the function of sE-cad, we found that immunodepletion of sE-cad from the media of A549 cells untreated or treated with BDNF, nicotine, or epinephrine reduced activation of EGFR and IGF-1R, decreased PI3K and ERK1/2 activities, increased p53 activation, decreased cell viability, and increased apoptosis, while no effects were found using H1299 cells under all conditions tested.

5.
Cells ; 12(3)2023 01 25.
Article in English | MEDLINE | ID: mdl-36766747

ABSTRACT

Cisplatin is a platinum agent used in the treatment of non-small cell lung cancer (NSCLC). Much remains unknown regarding the basic operative mechanisms underlying cisplatin resistance in NSCLC. In this study, we found that phosphorylation of IGFBP-3 by CK2 (P-IGFBP-3) decreased its binding to hyaluronan (HA) but not to IGF-1 and rendered the protein less effective at reducing cell viability or increasing apoptosis than the non-phosphorylated protein with or without cisplatin in the human NSCLC cell lines, A549 and H1299. Our data suggest that blocking CD44 signaling augmented the effects of cisplatin and that IGFBP-3 was more effective at inhibiting HA-CD44 signaling than P-IGFBP-3. Blocking CK2 activity and HA-CD44 signaling increased cisplatin sensitivity and more effectively blocked the PI3K and AKT activities and the phospho/total NFκB ratio and led to increased p53 activation in A549 cells. Increased cell sensitivity to cisplatin was observed upon co-treatment with inhibitors targeted against PI3K, AKT, and NFκB while blocking p53 activity decreased A549 cell sensitivity to cisplatin. Our findings shed light on a novel mechanism employed by CK2 in phosphorylating IGFBP-3 and increasing cisplatin resistance in NSCLC. Blocking phosphorylation of IGFBP-3 by CK2 may be an effective strategy to increase NSCLC sensitivity to cisplatin.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Cisplatin/metabolism , Casein Kinase II/metabolism , Phosphorylation , Hyaluronic Acid/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Cell Survival , Tumor Suppressor Protein p53/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Hyaluronan Receptors/metabolism
6.
Int J Mol Sci ; 23(21)2022 Oct 24.
Article in English | MEDLINE | ID: mdl-36361620

ABSTRACT

It is well-recognized that cigarette smoking is a primary risk factor in the development of non-small cell lung cancer (NSCLC), known to account for ~80% of all lung cancers with nicotine recognized as the major addictive component. In investigating the effect of nicotine, brain-derived neurotrophic factor (BDNF), and the ß-adrenergic receptor blocker, propranolol, on sensitivity of NSCLC cell lines, A549 and H1299, to cisplatin, we found increased cell viability, and enhanced cisplatin resistance with nicotine and/or BDNF treatment while opposite effects were found upon treatment with propranolol. Cell treatment with epinephrine or nicotine led to EGFR and IGF-1R activation, effects opposite to those found with propranolol. Blocking EGFR and IGF-1R activation increased cell sensitivity to cisplatin in both cell lines. PI3K and AKT activities were upregulated by nicotine or BDNF and downregulated by cell treatment with inhibitors against EGFR and IGF-1R and by propranolol. Apoptosis and cell sensitivity to cisplatin increased upon co-treatment of cells with cisplatin and inhibitors against PI3K or AKT. Our findings shed light on an interplay between nicotine, BDNF, and ß-Adrenergic receptor signaling in regulating survival of lung cancer cells and chemoresistance which can likely expand therapeutic opportunities that target this regulatory network in the future.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Nicotine/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , ErbB Receptors/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use , Adrenergic Antagonists/pharmacology , Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Adrenergic, beta , Cell Line, Tumor
7.
Cells ; 11(22)2022 11 08.
Article in English | MEDLINE | ID: mdl-36428962

ABSTRACT

In this study, we examined the roles of heparanase and IGFBP-3 in regulating A549 and H1299 non-small-cell lung cancer (NSCLC) survival. We found that H1299 cells, known to be p53-null with no expression of IGFBP-3, had higher heparanase levels and activity and higher levels of heparan sulfate (HS) in the media compared to the media of A549 cells. Inhibiting heparanase activity or its expression using siRNA had no effect on the levels of IGFBP-3 in the media of A549 cells, reduced the levels of soluble HS fragments, and led to decreased interactions between IGFBP-3 and HS in the media. HS competed with HA for binding to IGFBP-3 or IGFBP-3 peptide (215-KKGFYKKKQCRPSKGRKR-232) but not the mutant peptide (K228AR230A). HS abolished the cytotoxic effects of IGFBP-3 but not upon blocking HA-CD44 signaling with the anti-CD44 antibody (5F12). Blocking HA-CD44 signaling decreased the levels of heparanase in the media of both A549 and H1299 cell lines and increased p53 activity and the levels of IGFBP-3 in A549 cell media. Knockdown of p53 led to increased heparanase levels and reduced IGFBP-3 levels in A549 cell media while knockdown of IGFBP-3 in A549 cells blocked p53 activity and increased heparanase levels in the media.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , A549 Cells , Cell Survival , Heparitin Sulfate/metabolism , Insulin-Like Growth Factor Binding Protein 3 , Lung Neoplasms/metabolism , Peptides/metabolism , Tumor Suppressor Protein p53
8.
Int J Mol Sci ; 23(18)2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36142659

ABSTRACT

In comparing two human lung cancer cells, we previously found lower levels of acetylcholinesterase (AChE) and intact amyloid-ß40/42 (Aß), and higher levels of mature brain-derived neurotrophic factor (mBDNF) in the media of H1299 cells as compared to A549 cell media. In this study, we hypothesized that the levels of soluble amyloid precursor protein α (sAPPα) are regulated by AChE and mBDNF in A549 and H1299 cell media. The levels of sAPPα were higher in the media of H1299 cells. Knockdown of AChE led to increased sAPPα and mBDNF levels and correlated with decreased levels of intact Aß40/42 in A549 cell media. AChE and mBDNF had opposite effects on the levels of Aß and sAPPα and were found to operate through a mechanism involving α-secretase activity. Treatment with AChE decreased sAPPα levels and simultaneously increased the levels of intact Aß40/42 suggesting a role of the protein in shifting APP processing away from the non-amyloidogenic pathway and toward the amyloidogenic pathway, whereas treatment with mBDNF led to opposite effects on those levels. We also show that the levels of sAPPα are regulated by protein kinase C (PKC), extracellular signal-regulated kinase (ERK)1/2, phosphoinositide 3 Kinase (PI3K), but not by protein kinase A (PKA).


Subject(s)
Alzheimer Disease , Lung Neoplasms , Acetylcholinesterase , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain-Derived Neurotrophic Factor , Cyclic AMP-Dependent Protein Kinases , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Protein Kinase C/metabolism
9.
Proteins ; 90(3): 835-847, 2022 03.
Article in English | MEDLINE | ID: mdl-34766381

ABSTRACT

Ubiquitin-like containing PHD and ring finger (UHRF)1 and UHRF2 are multidomain epigenetic proteins that play a critical role in bridging crosstalk between histone modifications and DNA methylation. Both proteins contain two histone reader domains, called tandem Tudor domain (TTD) and plant homeodomain (PHD), which read the modification status on histone H3 to regulate DNA methylation and gene expression. To shed light on the mechanism of histone binding by UHRF2, we have undergone a detailed molecular investigation with the TTD, PHD and TTD-PHD domains and compared the binding activity to its UHRF1 counterpart. We found that unlike UHRF1 where the PHD is the primary binding contributor, the TTD of UHRF2 has modestly higher affinity toward the H3 tail, while the PHD has a weaker binding interaction. We also demonstrated that like UHRF1, the aromatic amino acids within the TTD are important for binding to H3K9me3 and a conserved aspartic acid within the PHD forms an ionic interaction with R2 of H3. However, while the aromatic amino acids in the TTD of UHRF1 contribute to selectivity, the analogous residues in UHRF2 contribute to both selectivity and affinity. We also discovered that the PHD of UHRF2 contains a distinct asparagine in the H3R2 binding pocket that lowers the binding affinity of the PHD by reducing a potential electrostatic interaction with the H3 tail. Furthermore, we demonstrate the PHD and TTD of UHRF2 cooperate to interact with the H3 tail and that dual domain engagement with the H3 tail relies on specific amino acids. Lastly, our data indicate that the unique stretch region in the TTD of UHRF2 can decrease the melting temperature of the TTD-PHD and represents a disordered region. Thus, these subtle but important mechanistic differences are potential avenues for selectively targeting the histone binding interactions of UHRF1 and UHRF2 with small molecules.


Subject(s)
Histones/chemistry , Homeodomain Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Binding Sites , CCAAT-Enhancer-Binding Proteins/chemistry , DNA Methylation , Epigenesis, Genetic , Escherichia coli/genetics , Gene Expression , Humans , Protein Binding , Protein Processing, Post-Translational , Structure-Activity Relationship , Tudor Domain , Ubiquitin-Protein Ligases/genetics
10.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209215

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, has been linked to several human malignancies and shown to promote tumorigenesis. The purpose of this study was to explore the relative abundance of pro-brain-derived neurotrophic factor (proBDNF) and mature BDNF (mBDNF) in A549 (p53 wild-type) and H1299 (p53-null) lung cancer cell media. Higher levels of proBDNF were detected in the media of A549 cells than in H1299 cell media. Using inhibitors, we found that the levels of proBDNF and mBDNF in the media are likely regulated by PI3K, AKT, and NFκB. However, the largest change in these levels resulted from MMP2/9 inhibition. Blocking p53 function in A549 cells resulted in increased mBDNF and decreased proBDNF, suggesting a role for p53 in regulating these levels. The ratio of proBDNF/mBDNF was not affected by MMP2 knockdown but increased in the media of both cell lines upon knockdown of MMP9. Downregulation of either MMP2 or MMP9 by siRNA showed that MMP9 siRNA treatment of either A549 or H1299 cells resulted in decreased cell viability and increased apoptosis, an effect diminished upon the same treatment with proBDNF immunodepleted media, suggesting that MMP9 regulates the cytotoxic effects induced by proBDNF in lung cancer cells.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Culture Media/metabolism , Lung Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Up-Regulation , A549 Cells , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mutation , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
11.
Sci Rep ; 11(1): 9708, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33958632

ABSTRACT

In this study, we set out to identify regulators of intact amyloid-ß40/42 (Aß) levels in A549 (p53 wild-type) and H1299 (p53-null) lung cancer cell media. Higher Aß levels were detected in the media of A549 than H1299 cells without or with treatment with 4-methylumbelliferone (4-MU) and/or the anti-CD44 antibody (5F12). Using inhibitors, we found that PI3K, AKT, and NFκB are likely involved in regulating Aß levels in the media. However, increased Aß levels that more closely resembled those found upon 4-MU co-treatment resulted from MMP2/9 inhibition, suggesting that MMP2/9 maybe the main contributors to regulation of Aß levels in the media. Differences in Aß levels might be accounted for, in part, by p53 since blocking p53 function in A549 cells resulted in decreased Aß levels, increased MMP2/9 levels, increased PI3K/AKT activities and the phospho/total NFκB ratio. Using siRNA targeted against MMP2 or MMP9, we found increased Aß levels in the media, however, MMP2 knockdown led to Aß levels closely mimicking those detected by co-treatment with 4-MU. Cell viability or apoptosis upon treatment with either MMP2 or MMP9 siRNA along with Aß immunodepletion, showed that MMP2 is the predominant regulator of the cytotoxic effects induced by Aß in lung cancer cells.


Subject(s)
Amyloid beta-Peptides/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Benzothiazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Culture Media, Conditioned , Humans , Hyaluronan Receptors/immunology , Hymecromone/pharmacology , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Toluene/analogs & derivatives , Toluene/pharmacology
12.
FEBS Open Bio ; 10(12): 2805-2823, 2020 12.
Article in English | MEDLINE | ID: mdl-33145964

ABSTRACT

Humanin (HN) is known to bind amyloid beta (Aß)-inducing cytoprotective effects, while binding of acetylcholinesterase (AChE) to Aß increases its aggregation and cytotoxicity. Previously, we showed that binding of HN to Aß blocks aggregation induced by AChE and that HN decreases but does not abolish Aß-AChE interactions in A549 cell media. Here, we set out to shed light on factors that modulate the interactions of Aß with HN and AChE. We found that binding of either HN or AChE to Aß is not affected by heparan sulfate, while ATP, thought to reduce misfolding of Aß, weakened interactions between AChE and Aß but strengthened those between Aß and HN. Using media from either A549 or H1299 lung cancer cells, we observed that more HN was bound to Aß upon addition of ATP, while levels of AChE in a complex with Aß were decreased by ATP addition to A549 cell media. Exogenous addition of ATP to either A549 or H1299 cell media increased interactions of endogenous HN with Aß to a comparable extent despite differences in AChE expression in the two cell lines, and this was correlated with decreased binding of exogenously added HN to Aß. Treatment with exogenous ATP had no effect on cell viability under all conditions examined. Exogenously added ATP did not affect viability of cells treated with AChE-immunodepleted media, and there was no apparent protection against the cytotoxicity resulting from immunodepletion of HN. Moreover, exogenously added ATP had no effect on the relative abundance of oligomer versus total Aß in either cell line.


Subject(s)
Acetylcholinesterase/metabolism , Adenosine Triphosphate/metabolism , Amyloid beta-Peptides/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Humans , Tumor Cells, Cultured
13.
FEBS Open Bio ; 10(8): 1668-1684, 2020 08.
Article in English | MEDLINE | ID: mdl-32592613

ABSTRACT

IGFBP-3, the most abundant IGFBP and the main carrier of insulin-like growth factor I (IGF-I) in the circulation, can bind IGF-1 with high affinity, which attenuates IGF/IGF-IR interactions, thereby resulting in antiproliferative effects. The C-terminal domain of insulin-like growth factor-binding protein-3 (IGFBP-3) is known to contain an 18-basic amino acid motif capable of interacting with either humanin (HN) or hyaluronan (HA). We previously showed that the 18-amino acid IGFBP-3 peptide is capable of binding either HA or HN with comparable affinities to the full-length IGFBP-3 protein and that IGFBP-3 can compete with the HA receptor, CD44, for binding HA. Blocking the interaction between HA and CD44 reduced viability of A549 human lung cancer cells. In this study, we set out to better characterize IGFBP-3-HA interactions. We show that both stereochemistry and amino acid identity are important determinants of the interaction between the IGFBP-3 peptide and HA and for the peptide's ability to exert its cytotoxic effects. Binding of IGFBP-3 to either HA or HN was unaffected by glycosylation or reduction of IGFBP-3, suggesting that the basic 18-amino acid residue sequence of IGFBP-3 remains accessible for interaction with either HN or HA upon glycosylation or reduction of the full-length protein. Removing N-linked oligosaccharides from CD44 increased its ability to compete with IGFBP-3 for binding HA, while reduction of CD44 rendered the protein relatively ineffective at blocking IGFBP-3-HA interactions. We conclude that both deglycosylation and disulfide bond formation are important for CD44 to compete with IGFBP-3 for binding HA.


Subject(s)
Hyaluronic Acid/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , A549 Cells , Disulfides/chemistry , Disulfides/metabolism , Glycosylation , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Insulin-Like Growth Factor Binding Protein 3/chemistry , Intracellular Signaling Peptides and Proteins/chemistry
14.
Biochemistry ; 59(21): 1981-2002, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32383868

ABSTRACT

It is known that the humanin (HN) peptide binding to amyloid-ß (Aß) protects against its cytotoxic effects, while acetylcholinesterase (AChE) binding to Aß increases its aggregation and cytotoxicity. HN is also known to bind the insulin-like growth factor binding protein-3 (IGFBP-3). Here, we examined the regulation of Aß conformations by HN, AChE, and IGFBP-3 both in vitro and in the conditioned media from A549 and H1299 lung cancer cells. Our in vitro results showed the following: IGFBP-3 binds HN and blocks it from binding Aß in the absence or presence of AChE; HN and AChE can simultaneously bind Aß but not when in the presence of IGFBP-3; HN is unable to reduce the aggregation of Aß in the presence of IGFBP-3; and HN abolishes the aggregation of Aß induced by the addition of AChE in the absence of IGFBP-3. In the media, AChE and HN can simultaneously bind Aß. While both AChE and HN are detected when using 6E10 Aß antibodies, only AChE is detected when using the Aß 17-24 antibody 4G8, the anti-oligomer A11, and the anti-amyloid fibril LOC antibodies. No signal was observed for IGFBP-3 with any of the anti-amyloid antibodies used. Exogenously added IGFBP-3 reduced the amount of HN found in a complex when using 6E10 antibodies and correlated with a concomitant increase in the amyloid oligomers. Immunodepletion of HN from the media of the A549 and H1299 cells increased the relative abundance of the oligomer vs the total amount of Aß, the A11-positive prefibrillar oligomers, and to a lesser extent the LOC-positive fibrillar oligomers, and was also correlated with diminished cell viability and increased apoptosis.


Subject(s)
Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Humans , Protein Aggregates , Tumor Cells, Cultured
15.
Sci Rep ; 10(1): 5083, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32193421

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) belongs to a family of six IGF binding proteins. We previously found that IGFBP-3 exerts its cytotoxic effects on A549 (p53 wild-type) cell survival through a mechanism that depends on hyaluronan-CD44 interactions. To shed light on the mechanism employed, we used CD44-negative normal human lung cells (HFL1), A549, and H1299 (p53-null) lung cancer cells. A synthetic IGFBP-3 peptide (215-KKGFYKKKQCRPSKGRKR-232) but not the mutant (K228AR230A), was able to bind hyaluronan more efficiently than the analogous sequences from the other IGFBPs. In a manner comparable to that of the IGFBP-3 protein, the peptide blocked hyaluronan-CD44 signaling, and more effectively inhibited viability of A549 cells than viability of either H1299 or HFL1 cell lines. Treatment with the IGFBP-3 protein or its peptide resulted in increased acetylcholinesterase concentration and activity in the A549 cell media but not in the media of either HFL1 or H1299, an effect that correlated with increased apoptosis and decreased cell viability. These effects were diminished upon the same treatment of A549 cells transfected with either p53 siRNA or acetylcholinesterase siRNA. Taken together, our results show that IGFBP-3 or its peptide blocks hyaluronan-CD44 signaling via a mechanism that depends on both p53 and acetylcholinesterase.


Subject(s)
Acetylcholinesterase/metabolism , Apoptosis/genetics , Culture Media/metabolism , Hyaluronan Receptors/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor Binding Protein 3/physiology , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , A549 Cells , Apoptosis/drug effects , Cell Survival/drug effects , Humans , Hyaluronic Acid/metabolism , Signal Transduction/drug effects
16.
Protein Pept Lett ; 26(7): 502-511, 2019.
Article in English | MEDLINE | ID: mdl-30950343

ABSTRACT

BACKGROUND: Amyloid fibrils in Alzheimer's disease are composed of amyloid-ß (Aß) peptides of variant lengths. Humanin (HN), a 24 amino acid residue neuroprotective peptide, is known to interact with the predominant Aß isoform in the brain, Aß (1-40). METHODS: Here, we constructed smaller segments of Aß and HN and identified residues in HN important for both HN-HN and HN-Aß interactions. Peptides corresponding to amino acid residues 5- 15 of HN, HN (5-15), HN (5-15, L11S), where Leu11 was replaced with Ser, and residues 17-28 of Aß, Aß (17-28), were synthesized and tested for their ability to block formation of the complex between HN and Aß (1-40). RESULTS: Co-immunoprecipitation and binding kinetics showed that HN (5-15) was more efficient at blocking the complex between HN and Aß (1-40) than either HN (5-15, L11S) or Aß (17-28). Binding kinetics of these smaller peptides with either full-length HN or Aß (1-40) showed that HN (5- 15) was able to bind either Aß (1-40) or HN more efficiently than HN (5-15, L11S) or Aß (17-28). Compared to full-length HN, however, HN (5-15) bound Aß (1-40) with a weaker affinity suggesting that while HN (5-15) binds Aß, other residues in the full length HN peptide are necessary for maximum interactions. CONCLUSION: L11 was more important for interactions with Aß (1-40) than with HN. Aß (17-28) was relatively ineffective at binding to either Aß (1-40) or HN. Moreover, HN, and the smaller HN (5-15), HN (5-15 L11S), and Aß (17-28) peptides, had different effects on regulating Aß (1-40) aggregation kinetics.


Subject(s)
Amyloid beta-Peptides/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Humans , Kinetics , Protein Conformation
17.
FEBS Lett ; 593(7): 732-742, 2019 04.
Article in English | MEDLINE | ID: mdl-30821397

ABSTRACT

Dysfunction of the dopaminergic pathway is linked to numerous diseases of the nervous system. The D1-D2 receptor heteromer is known to play a role in certain neuropsychiatric disorders, such as depression. Here, we synthesized an eight amino acid residue peptide, EAARRAQE, derived from the third intracellular loop of the D2 receptor and show that the peptide binds to the D1 receptor with comparable efficiency as that of the full-length D2 receptor protein. Moreover, immunoprecipitation studies show the existence of a heteromeric complex formed both in vitro and in total protein derived from temporal and frontal lobe tissue from normal and depressed subjects. The efficiency of the peptide to block the D1-D2 heteromeric complex was comparable in all the samples tested.


Subject(s)
Dopamine D2 Receptor Antagonists/pharmacology , Neurons/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Animals , Brain Mapping , Depression/genetics , Depression/pathology , Dopamine/genetics , Dopamine/metabolism , Dopamine D2 Receptor Antagonists/chemical synthesis , Frontal Lobe/metabolism , Humans , Immunoprecipitation , Neurons/drug effects , Neurons/pathology , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding/genetics , Rats , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D2/chemistry , Temporal Lobe/metabolism
18.
Biochemistry ; 57(39): 5726-5737, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30184438

ABSTRACT

Insulin-like growth factor-binding protein-3 (IGFBP-3) belongs to a family of IGF-binding proteins. Humanin is a peptide known to bind residues 215-232 of mature IGFBP-3 in the C-terminal region of the protein. This region of IGFBP-3 was shown earlier to bind certain glycosaminoglycans including hyaluronan (HA). Here, we characterized the binding affinities of the IGFBP-3 protein and peptide (215-KKGFYKKKQCRPSKGRKR-232) to HA and to humanin and found that HA binds with a weaker affinity to this region than does humanin. Either HA or humanin could bind to this IGFBP-3 segment, but not simultaneously. The HA receptor, CD44, blocked HA binding to IGFBP-3 but had no effect on binding of humanin to either IGFBP-3 or its peptide. Upon incubation of HA with CD44 and either IGFBP-3 protein or peptide, humanin was effective at binding and sequestering IGFBP-3 or peptide, thereby enabling access of CD44 to HA. We show that IGFBP-3 and humanin in the medium of A549 lung cancer cells can immunoprecipitate in a complex. However, the fraction of IGFBP-3 in the medium that is able to bind HA was not complexed with humanin suggesting that HA binding to the 215-232 segment renders it inaccessible for binding to humanin. Moreover, while the cytotoxic effects of IGFBP-3 on cell viability were reversed by humanin, blocking HA-CD44 interaction with an anti-CD44 antibody in combination with IGFBP-3 did not have an additive negative effect on cell viability suggesting that IGFBP-3 exerts its cytotoxic effects on cell survival through a mechanism that depends on HA-CD44 interactions.


Subject(s)
Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , A549 Cells , Amino Acid Sequence , Cell Survival/physiology , Humans , Peptide Fragments/metabolism , Protein Binding
19.
Protein Pept Lett ; 24(7): 590-598, 2017.
Article in English | MEDLINE | ID: mdl-28641565

ABSTRACT

INTRODUCTION: The importance of the antitumor activity of some antimicrobial peptides (AMPs) is being increasingly recognized. The antimicrobial peptide, tachyplesin, has been shown to exhibit anticancer properties and a linear, cysteine deleted analogue (CDT), was found to retain its antibacterial function. OBJECTIVES: The objective was to test CDT and related analogues against normal mammalian, bacterial, and cancer cells to determine their effectiveness and then utilize specific assays to determine a possible mechanism of action. METHODS: We used sequence reversal and D-amino acids to synthesize four CDT analogues by solid phase peptide synthesis. A number of assays were used including liposome dye-leakage, antibacterial activity against both Gram-positive and Gram-negative bacterial strains, hemolytic assays, methyl thiazolyl tetrazolium (MTT), and apoptosis to examine their effectiveness as both AMPs and anti-cancer peptides (ACPs). We then tested the analogues for their ability to inhibit proliferation of the human lung cancer cell line, A549. RESULTS: We found that D-CDT exhibited the best bactericidal properties of those tested and was not damaging to red blood cells. Both D-CDT and reverse D-CDT showed a dose-dependent reduction of cell viability. However, D-CDT was most effective with an IC50 of 9.814 µM, a value 9-fold lower than that calculated for reverse D-CDT (90.16 µM). Apoptosis does not appear to be a mechanism by which D-CDT exerts its anticancer properties since > 100 µM was required to increase activation of caspase 3. Moreover, the ERK1/2 pathway is also unlikely since only a modest (20%) decrease of activity was observed with > 100 µM D-CDT. However, D-CDT was found to operate via a hyaluronan (HA)-dependent mechanism as pretreatment of the cells with hyaluronidase decreased the cytotoxic effects of D-CDT on A549 cells and increased its IC50 29-fold to 283.9 µM. CONCLUSION: D-CDT is both an effective AMP and ACP, and likely exerts its anticancer effects through both membranolytic as well as an HA-mediated mechanism.


Subject(s)
Adenocarcinoma/drug therapy , Antimicrobial Cationic Peptides/administration & dosage , Cell Proliferation/drug effects , DNA-Binding Proteins/administration & dosage , Lung Neoplasms/drug therapy , Peptides, Cyclic/administration & dosage , A549 Cells , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Amino Acid Sequence , Amino Acids/administration & dosage , Amino Acids/chemistry , Antimicrobial Cationic Peptides/chemistry , Apoptosis/drug effects , Cysteine/chemistry , Drug Screening Assays, Antitumor , Gram-Negative Bacteria/drug effects , Humans , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects
20.
Protein Pept Lett ; 22(10): 869-76, 2015.
Article in English | MEDLINE | ID: mdl-26216267

ABSTRACT

Nuclear translocation of IGFBP3 by importin-ß1 is a prerequisite for IGFBP3-induced apoptosis. The neuroprotective peptide humanin (HN) counteracts IGFBP3-induced cell death. However, the mechanism by which humanin protects cells is currently unknown. The natural synthesis of this peptide decreases with age, coincident with the likelihood for the development of Alzheimer's Disease, making it a promising target for therapeutics. We have examined the effect of full-length humanin and a synthetic analogue (HN 3-19), known to be sufficient for its neuroprotective function, on the interaction between IGFBP3 and importin-ß1. Using competitive ligand dot blotting, co-immunoprecipitation, and an ELISA-based binding assay, we determined that 1) humanin binds to IGFBP3 with a Kd of 5.05 µM and 2) both humanin (IC50 of 18.1 µM) and HN 3-19 (IC50 of 10.3 µM) interfere with the binding of importin-ß1 to IGFBP3 in vitro. We also demonstrated that HN 3-19 is able to reduce the rate of apoptosis in a human lung adenocarcinoma cell line, suggesting a possible mechanism of action for humanin as an inhibitor of IGFBP3 nuclear translocation. Understanding the exact mechanism by which humanin and its analogue, HN 3-19, bind to IGFPB3 and regulate its interaction with importin-ß1 will open the door to modulating the protein-protein interactions involved in neuronal cell death.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/chemistry , Peptides/chemistry , beta Karyopherins/chemistry , Cell Line, Tumor , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Intracellular Signaling Peptides and Proteins , Peptides/metabolism , Protein Binding , beta Karyopherins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...