Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Pharm ; 20(9): 4559-4573, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37555521

ABSTRACT

The enzyme PACE4 has been validated as a promising therapeutic target to expand the range of prostate cancer (PCa) treatments. In recent years, we have developed a potent peptidomimetic inhibitor, namely, compound C23 (Ac-(DLeu)LLLRVK-4-amidinobenzylamide). Like many peptides, C23 suffers from an unfavorable drug-like profile which, despite our efforts, has not yet benefited from the usual SAR studies. Hence, we turned our attention toward a novel formulation strategy, i.e., the use of cyclodextrins (CDs). CDs can benefit compounds through the formation of "host-guest" complexes, shielding the guest from degradation and enhancing biological survival. In this study, a series of ßCD-C23 complexes have been generated and their properties evaluated, including potency toward the enzyme in vitro, a cell-based proliferation assay, and stability in plasma. As a result, a new ßCD-formulated lead compound has been identified, which, in addition to being more soluble and more potent, also showed an improved stability profile.


Subject(s)
Cyclodextrins , beta-Cyclodextrins , Male , Humans , Peptides/pharmacology , beta-Cyclodextrins/pharmacology , Cyclodextrins/pharmacology , Cyclodextrins/chemistry
2.
Can J Physiol Pharmacol ; 98(7): 477-481, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32119574

ABSTRACT

Recent studies in prostate cancer have identified PACE4, a proprotein convertase enzyme, as an emerging therapeutic target. Inhibition of PACE4-altCT, an oncogenic isoform of PACE4, using molecular or pharmacological approaches results in decreased cell proliferation and tumor progression in xenograft models. Although several validations have confirmed PACE4-altCT as a novel therapeutic target, the transcriptional regulation of PACE4 isoforms and mechanism of action remain a challenge. Previously, it has been reported that the human PACE4 promoter possesses potential binding sites for the E2F family of transcription factors, all of which are involved in cell cycle regulation and synthesis of DNA in mammalian cells. Therefore, we attempted to conduct in-depth evaluation of E2Fs on PACE4 and PACE4 isoform expression in prostate cancer. We conducted in vitro molecular silencing studies in various prostate cancer cell lines and determined the change in PACE4 expression levels. The results clearly show that the E2Fs alone do not alter PACE4 expression.


Subject(s)
Adenocarcinoma/genetics , E2F Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Proprotein Convertases/genetics , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Datasets as Topic , E2F Transcription Factors/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Male , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Up-Regulation
3.
J Biol Chem ; 292(25): 10564-10573, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28468828

ABSTRACT

Protein C, a secretory vitamin K-dependent anticoagulant serine protease, inactivates factors Va/VIIIa. It is exclusively synthesized in liver hepatocytes as an inactive zymogen (proprotein C). In humans, thrombin cleavage of the propeptide at PR221↓ results in activated protein C (APC; residues 222-461). However, the propeptide is also cleaved by a furin-like proprotein convertase(s) (PCs) at KKRSHLKR199↓ (underlined basic residues critical for the recognition by PCs), but the order of cleavage is unknown. Herein, we present evidence that at the surface of COS-1 cells, mouse proprotein C is first cleaved by the convertases furin, PC5/6A, and PACE4. In mice, this cleavage occurs at the equivalent site, KKRKILKR198↓, and requires the presence of Arg198 at P1 and a combination of two other basic residues at either P2 (Lys197), P6 (Arg193), or P8 (Lys191) positions. Notably, the thrombin-resistant R221A mutant is still cleaved by these PCs, revealing that convertase cleavage can precede thrombin activation. This conclusion was supported by the fact that the APC-specific activity in the medium of COS-1 cells is exclusively dependent on prior cleavage by the convertases, because both R198A and R221A lack protein C activity. Primary cultures of hepatocytes derived from wild-type or hepatocyte-specific furin, PC5/6, or complete PACE4 knock-out mice suggested that the cleavage of overexpressed proprotein C is predominantly performed by furin intracellularly and by all three proprotein convertases at the cell surface. Indeed, plasma analyses of single-proprotein convertase-knock-out mice showed that loss of the convertase furin or PC5/6 in hepatocytes results in a ∼30% decrease in APC levels, with no significant contribution from PACE4. We conclude that prior convertase cleavage of protein C in hepatocytes is critical for its thrombin activation.


Subject(s)
Hepatocytes/enzymology , Liver/enzymology , Proprotein Convertase 5/metabolism , Protein C/metabolism , Amino Acid Substitution , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/physiology , Hep G2 Cells , Humans , Mice , Mice, Knockout , Mutation, Missense , Proprotein Convertase 5/genetics , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Protein C/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Thrombin/genetics , Thrombin/metabolism
4.
J Biol Chem ; 291(47): 24676-24687, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27758865

ABSTRACT

The mechanism of LDL receptor (LDLR) degradation mediated by the proprotein convertase subtilisin/kexin type 9 (PCSK9) has been extensively studied; however, many steps within this process remain unclear and still require characterization. Recent studies have shown that PCSK9 lacking its Cys/His-rich domain can still promote LDLR internalization, but the complex does not reach the lysosome suggesting the presence of an additional interaction partner(s). In this study we carried out an unbiased screening approach to identify PCSK9-interacting proteins in the HepG2 cells' secretome using co-immunoprecipitation combined with mass spectrometry analyses. Several interacting proteins were identified, including glypican-3 (GPC3), phospholipid transfer protein, matrilin-3, tissue factor pathway inhibitor, fibrinogen-like 1, and plasminogen activator inhibitor-1. We then validated these interactions by co-immunoprecipitation and Western blotting. Furthermore, functional validation was examined by silencing each candidate protein in HepG2 cells using short hairpin RNAs to determine their effect on LDL uptake and LDLR levels. Only GPC3 and phospholipid transfer protein silencing in HepG2 cells significantly increased LDL uptake in these cells and displayed higher total LDLR protein levels compared with control cells. Moreover, our study provides the first evidence that GPC3 can modulate the PCSK9 extracellular activity as a competitive binding partner to the LDLR in HepG2 cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glypicans/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Carcinoma, Hepatocellular/genetics , Glypicans/genetics , Hep G2 Cells , Humans , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Liver Neoplasms/genetics , Matrilin Proteins/genetics , Matrilin Proteins/metabolism , Neoplasm Proteins/genetics , Proprotein Convertase 9/genetics , Protein Binding , Receptors, LDL/genetics
5.
Proc Natl Acad Sci U S A ; 110(43): 17362-7, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101515

ABSTRACT

PC7 belongs to the proprotein convertase family, whose members are implicated in the cleavage of secretory precursors. The in vivo function of PC7 is unknown. Herein, we find that the precursor proBDNF is processed into mature BDNF in COS-1 cells coexpressing proBDNF with either PC7 or Furin. Conversely, the processing of proBDNF into BDNF is markedly reduced in the absence of either Furin or PC7 in mouse primary hepatocytes. In vivo we observe that BDNF and PC7 mRNAs are colocalized in mouse hippocampus and amygdala and that mature BDNF protein levels are reduced in these brain areas in PC7 KO mice but not in the hippocampus of PC1/3 KO mice. Various behavioral tests reveal that in PC7 KO mice spatial memory is intact and plasticity of responding is mildly abnormal. Episodic and emotional memories are severely impaired, but both are rescued with the tyrosine receptor kinase B agonist 7,8-dihydroxyflavone. Altogether, these results support an in vivo role for PC7 in the regulation of certain types of cognitive performance, in part via proBDNF processing. Because polymorphic variants of human PC7 are being characterized, it will be important in future studies to determine their effects on additional physiological and behavioral processes.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Maze Learning/physiology , Memory/physiology , Subtilisins/metabolism , Amygdala/metabolism , Animals , Blotting, Western , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Flavanones/pharmacology , Gene Expression , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Hippocampus/metabolism , Humans , In Situ Hybridization , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Knockout , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subtilisins/genetics
6.
J Biol Chem ; 289(25): 17732-46, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24808179

ABSTRACT

Annexin A2 (AnxA2) was reported to be an extracellular endogenous inhibitor of proprotein convertase subtilisin kexin type 9 (PCSK9) activity on cell-surface LDL receptor degradation. In this study, we investigated the effect of silencing the expression of AnxA2 and PCSK9 in HepG2 and Huh7 cells to better define the role of AnxA2 in PCSK9 regulation. AnxA2 knockdown in Huh7 cells significantly increased PCSK9 protein levels as opposed to AnxA2 knockdown in HepG2 cells. However, HepG2 cells overexpressing AnxA2 had lower levels of PCSK9 protein. Overall, our data revealed a plausible new role of AnxA2 in the reduction of PCSK9 protein levels via a translational mechanism. Moreover, the C-terminal Cys/His-rich domain of PCSK9 is crucial in the regulation of PCSK9 activity, and we demonstrated by far-Western blot assay that the M1 and M2 domains are necessary for the specific interaction of PCSK9's C-terminal Cys/His-rich domain and AnxA2. Finally, we produced and purified recombinant PCSK9 from humans and mice, which was characterized and used to perform 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate LDL cell-based assays on the stable knockdown HepG2 and Huh7 cells. We also demonstrated for the first time the equipotency of human and mouse PCSK9 R218S on human cells.


Subject(s)
Annexin A2/metabolism , Proprotein Convertases/biosynthesis , Protein Biosynthesis/physiology , Serine Endopeptidases/biosynthesis , Animals , Annexin A2/chemistry , Annexin A2/genetics , Gene Knockdown Techniques , HEK293 Cells , Hep G2 Cells , Humans , Mice , Proprotein Convertase 9 , Proprotein Convertases/chemistry , Proprotein Convertases/genetics , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
7.
J Biol Chem ; 287(18): 14703-17, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22396549

ABSTRACT

The proprotein convertase 1/3 is expressed in the regulated secretory pathway of neural and endocrine cells. Its major function is in the post-translational processing and activation of precursor proteins. The PC1/3 knock-out (KO) mouse model has allowed us to elucidate its physiological functions in studies focused primarily on neuroendocrine tissues. However, PC1/3 is also expressed in cells of the immune system, mainly in macrophages. The present study explores the effects of innate immune challenge in the PC1/3 KO mouse. PC1/3 KO mice have an enlarged spleen with marked disorganization of the marginal zone and red pulp. Immunohistochemical studies using various markers demonstrate a depletion of dendritic cells in PC1/3 KO spleens. When challenged with lipopolysaccharide, PC1/3 KO mice are more susceptible to septic shock than wild-type controls or other PC KO mice, such as PC2 and PC7 null mice. Plasma levels of proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) were very significantly elevated in PC1/3 KO mice, consistent with a hypercytokinemia, i.e. indicative of a major systemic uncontrolled inflammatory response or cytokine storm. Peritoneal macrophages isolated from PC1/3 KO mice also demonstrate elevated cytokine secretion when treated with LPS. Electron micrographs show morphological features indicating a prolonged activation of these cells following LPS stimulation. We also present evidence that the proinflammatory T(h)1 pathway is dominant in the PC1/3 KO mouse model. We conclude that aside from its important role in neuroendocrine functions PC1/3 also has an important role in the regulation of the innate immune system, most likely through the regulation of cytokine secretion in macrophages.


Subject(s)
Cytokines/immunology , Gene Expression Regulation, Enzymologic/immunology , Immunity, Innate , Macrophages, Peritoneal/immunology , Proprotein Convertase 1/immunology , Animals , Cytokines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/metabolism , Immune System Diseases/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/ultrastructure , Mice , Mice, Knockout , Proprotein Convertase 1/biosynthesis , Proprotein Convertase 1/genetics , Th1 Cells/enzymology , Th1 Cells/metabolism
8.
Sci Rep ; 12(1): 17489, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261691

ABSTRACT

Prostate cancer (PCa) is a complex disease progressing from in situ to invasive or metastatic tumors while also being capable of modulating its androgen dependence. Understanding how novel therapies are working across the different stages of the disease is critical for their proper positioning in the spectrum of PCa treatments. The targeting of proprotein convertase PACE4 (Paired basic Amino Acid-Cleaving Enzyme 4) has been proposed as a novel approach to treat PCa. Animal studies performed on LNCaP xenografts, an androgen-dependent model, already yielded positive results. In this study, we tested PACE4 inhibition on JHU-LNCaP-SM, a newly described androgen-independent model, in cell-based and xenograft assays. Like LNCaP, JHU-LNCaP-SM cells express PACE4 and its oncogenic isoform PACE4-altCT. Using isoform-specific siRNAs, downregulation of PACE4-altCT resulted in JHU-LNCaP-SM growth inhibition. Furthermore, JHU-LNCaP-SM responded to the PACE4 pharmacological inhibitor known as C23 in cell-based assays as well as in athymic nude mice xenografts. These data support the efficacy of PACE4 inhibitors against androgen independent PCa thereby demonstrating that PACE4 is a key target in PCa. The JHU-LNCaP-SM cell line represents a model featuring important aspects of androgen-independent PCa, but it also represents a very convenient model as opposed to LNCaP cells for in vivo studies, as it allows rapid screening due to its high implantation rate and growth characteristics as xenografts.


Subject(s)
Androgens , Prostatic Neoplasms , Mice , Animals , Male , Humans , Androgens/metabolism , Mice, Nude , Cell Line, Tumor , Prostatic Neoplasms/pathology , Proprotein Convertases/metabolism , Protein Isoforms , Amino Acids, Basic , Cell Proliferation , Receptors, Androgen
9.
Exp Lung Res ; 37(4): 205-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21309733

ABSTRACT

Nearly 40 potassium channels have been described in respiratory epithelial cells. Of these are found several members of the 4-transmembrane domain, 2-pore K(+) channel family (K2P family), namely Twik-1 and -2, Trek-1 and -2, Task-2, -3, and -4, Thik-1, and KCNK7. The aim of this study was to verify whether the Twik-related acid-sensitive K(+) channel, subtype 1 (Task-1) (also known as KCNK3), is present in the laryngeal mucosa in the newborn lamb. Through the use of immunohistochemistry and nested polymerase chain reaction (PCR) amplification, results indicate that Task-1 protein and mRNA are present in the laryngeal mucosa, in both the ciliated, pseudostratified columnar (respiratory) epithelium and the nonkeratinized, stratified squamous epithelium. The complete ovine Task-1 protein sequence showed high homology levels with previously reported mouse, bovine, and human Task-1 sequences. This includes a complete homology for the C-terminal amino acid sequence, which is mandatory for protein trafficking to the cell membrane. These results represent the first demonstration that Task-1, a pH-sensitive channel responsible for setting membrane potential, is present in the laryngeal mucosa of a newborn mammal.


Subject(s)
Laryngeal Mucosa/chemistry , Nerve Tissue Proteins/analysis , Potassium Channels, Tandem Pore Domain/analysis , Sheep , Animals , Animals, Newborn , Hydrogen-Ion Concentration , Immunohistochemistry , Membrane Potentials , Nerve Tissue Proteins/genetics , Polymerase Chain Reaction , Potassium Channels, Tandem Pore Domain/genetics , RNA, Messenger/analysis
10.
J Biol Chem ; 284(42): 28856-64, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19635789

ABSTRACT

Elevated levels of plasma low density lipoprotein (LDL)-cholesterol, leading to familial hypercholesterolemia, are enhanced by mutations in at least three major genes, the LDL receptor (LDLR), its ligand apolipoprotein B, and the proprotein convertase PCSK9. Single point mutations in PCSK9 are associated with either hyper- or hypocholesterolemia. Accordingly, PCSK9 is an attractive target for treatment of dyslipidemia. PCSK9 binds the epidermal growth factor domain A (EGF-A) of the LDLR and directs it to endosomes/lysosomes for destruction. Although the mechanism by which PCSK9 regulates LDLR degradation is not fully resolved, it seems to involve both intracellular and extracellular pathways. Here, we show that clathrin light chain small interfering RNAs that block intracellular trafficking from the trans-Golgi network to lysosomes rapidly increased LDLR levels within HepG2 cells in a PCSK9-dependent fashion without affecting the ability of exogenous PCSK9 to enhance LDLR degradation. In contrast, blocking the extracellular LDLR endocytosis/degradation pathway by a 4-, 6-, or 24-h incubation of cells with Dynasore or an EGF-AB peptide or by knockdown of endogenous autosomal recessive hypercholesterolemia did not significantly affect LDLR levels. The present data from HepG2 cells and mouse primary hepatocytes favor a model whereby depending on the dose and/or incubation period, endogenous PCSK9 enhances the degradation of the LDLR both extra- and intracellularly. Therefore, targeting either pathway, or both, would be an effective method to reduce PCSK9 activity in the treatment of hypercholesterolemia and coronary heart disease.


Subject(s)
Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Animals , Clathrin Light Chains/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , Hepatocytes/metabolism , Humans , Hypercholesterolemia/metabolism , Lysosomes/metabolism , Mice , Proprotein Convertase 9 , Proprotein Convertases , Protein Structure, Tertiary , trans-Golgi Network/metabolism
11.
Sci Rep ; 9(1): 2118, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765725

ABSTRACT

The proprotein convertase PACE4 has been validated as a potential target to develop new therapeutic interventions in prostate cancer (PCa). So far, the most effective compound blocking the activity of this enzyme has been designed based on the structure of a small peptide Ac-LLLLRVKR-NH2 known as the Multi-Leu (ML) peptide. Optimization of this scaffold led to the synthesis of compound C23 (Ac-[DLeu]LLLRVK-amidinobenzylamide) with a potent in vivo inhibitory effect on the tumor growth. However, further developments of PACE4 inhibitors may require additional improvements to counter their rapid renal clearance and to increase their tumor targeting efficiency. Herein, we explored the transformation of the ML-peptide into an albumin-binding prodrug containing a tumor specific release mechanism based on the prostate-specific antigen. Our data confirms that intravenous treatment using the ML-peptide alone has little effect on tumor growth, whereas by using the ML-prodrug in LNCaP xenograft-bearing mice it was significantly reduced. Additionally, excellent in vivo stability and tumor-targeting efficiency was demonstrated using a radiolabelled version of this compound. Taken together, these results provide a solid foundation for further development of targeted PACE4 inhibition in PCa.


Subject(s)
Albumins/metabolism , Peptide Fragments/pharmacology , Prodrugs/pharmacology , Proprotein Convertases/metabolism , Prostatic Neoplasms/drug therapy , Serine Endopeptidases/metabolism , Albumins/chemistry , Animals , Apoptosis , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Proprotein Convertases/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Conformation , Serine Endopeptidases/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
J Med Chem ; 61(18): 8457-8467, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30180568

ABSTRACT

The serine protease, PACE4, is a proprotein convertase that plays a substantial role in malignancy of prostate cancer. Our initial selective PACE4 inhibitor (Ac-LLLLRVKR-NH2) has evolved to the current lead compound C23 (Ac-dLeu-LLLRVK-Amba), which is active both in vitro and in vivo. By screening natural residues, except Cys, in C-terminal P1' position, it was established that increasing hydrophobicity was improving cell permeability, which was directly translated into PCa cells antiproliferative activity. This cell antiproliferation enhancement seems independent from effect of P1' residue on PACE4 affinity. Replacement of P1-Amba of C23 by Acpa (( S)-2-amino-3-(4-carbamimidoylphenyl)propanoic acid) followed by addition of tryptamine in P1' resulted in compound 32 exhibiting superior PCa cells antiproliferative activity over the reference compound C23 (3-fold). This study sheds light on key factors that improve cell penetrating property and antiproliferative activity of PACE4 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Proprotein Convertases/antagonists & inhibitors , Prostatic Neoplasms/pathology , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Male , Models, Molecular , Molecular Structure , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein Conformation , Serine Endopeptidases , Structure-Activity Relationship , Tumor Cells, Cultured
13.
J Med Chem ; 61(24): 11250-11260, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30501188

ABSTRACT

Paired basic amino acid cleaving enzyme 4 (PACE4), a serine endoprotease of the proprotein convertases family, has been recognized as a promising target for prostate cancer. We previously reported a selective and potent peptide-based inhibitor for PACE4, named the multi-Leu peptide (Ac-LLLLRVKR-NH2 sequence), which was then modified into a more potent and stable compound named C23 with the following structure: Ac-dLeu-LLLRVK-Amba (Amba: 4-amidinobenzylamide). Despite improvements in both in vitro and in vivo profiles of C23, its selectivity for PACE4 over furin was significantly reduced. We examined other Arg-mimetics instead of Amba to regain the lost selectivity. Our results indicated that the replacement of Amba with 5-(aminomethyl)picolinimidamide increased affinity for PACE4 and restored selectivity. Our results also provide a better insight on how structural differences between S1 pockets of PACE4 and furin could be employed in the rational design of selective inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Proprotein Convertases/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Humans , Male , Molecular Docking Simulation , Proprotein Convertases/chemistry , Proprotein Convertases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship
14.
J Endocrinol ; 192(3): 515-25, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17332521

ABSTRACT

We have previously shown that 7B2 null mice on the 129/SvEvTac (129) genetic background die at 5 weeks of age with hypercorticosteronemia due to a Cushing's-like disease unless they are rescued by adrenalectomy; however, 7B2 nulls on the C57BL/6NTac (B6) background remain healthy, with normal steroid levels. Since background exerts such a profound influence on the phenotype of this mutation, we have evaluated whether these two different mouse strains respond differently to high circulating steroids by chronically treating wild-type 129 and B6 mice with the synthetic steroid dexamethasone (Dex). Dex treatment decreased the dopamine content of the neurointermediate lobes (NIL) of 129 mice, leading to NIL enlargement and increased total D(2)R mRNA in the 129, but not the B6, NIL. Despite the decrease in this inhibitory transmitter, Dex-treated 129 mice exhibited reduced circulating alpha-melanocyte-stimulating hormone (alpha-MSH) along with reduced POMC-derived peptides compared with controls, possibly due to reduced POMC content in the NIL. In contrast, Dex-treated B6 mice showed lowered cellular ACTH, unchanged alpha-MSH and beta-endorphin, and increased circulating alpha-MSH, most likely due to increased cleavage of NIL ACTH by increased PC2. Dex-treated 129 mice exhibited hyperinsulinemia and lowered blood glucose, whereas Dex-treated B6 mice showed slightly increased glucose levels despite their considerably increased insulin levels. Taken together, our results suggest that the endocrinological response of 129 mice to chronic Dex treatment is very different from that of B6 mice. These strain-dependent differences in steroid sensitivity must be taken into account when comparing different lines of transgenic or knockout mice.


Subject(s)
Dexamethasone , Glucocorticoids , Pituitary Gland/drug effects , Animals , Blood Glucose/analysis , Blotting, Western/methods , Dopamine/blood , Glucagon/blood , In Situ Hybridization/methods , Insulin/blood , Melanocortins/blood , Melanotrophs/ultrastructure , Mice , Mice, Inbred Strains , Microscopy, Electron , Pituitary Gland/metabolism , Pituitary Gland/ultrastructure , Receptors, Dopamine D2/analysis , Species Specificity , beta-Endorphin/blood
15.
Mol Endocrinol ; 20(8): 1935-47, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16556729

ABSTRACT

Chromogranin A (CgA), originally identified in adrenal chromaffin cells, is a member of the granin family of acidic secretory glycoproteins that are expressed in endocrine cells and neurons. CgA has been proposed to play multiple roles in the secretory process. Intracellularly, CgA may control secretory granule biogenesis and target neurotransmitters and peptide hormones to granules of the regulated pathway. Extracellularly, peptides formed as a result of proteolytic processing of CgA may regulate hormone secretion. To investigate the role of CgA in the whole animal, we created a mouse mutant null for the Chga gene. These mice are viable and fertile and have no obvious developmental abnormalities, and their neural and endocrine functions are not grossly impaired. Their adrenal glands were structurally unremarkable, and morphometric analyses of chromaffin cells showed vesicle size and number to be normal. However, the excretion of epinephrine, norepinephrine, and dopamine was significantly elevated in the Chga null mutants. Adrenal medullary mRNA and protein levels of other dense-core secretory granule proteins including chromogranin B, and secretogranins II to VI were up-regulated 2- to 3-fold in the Chga null mutant mice. Hence, the increased expression of the other granin family members is likely to compensate for the Chga deficiency.


Subject(s)
Chromogranins/genetics , Chromogranins/physiology , Gene Targeting , Neurosecretory Systems/physiology , Secretory Vesicles/physiology , Adrenal Medulla/metabolism , Animals , Catecholamines/urine , Chromogranin A , Female , Gene Deletion , Gene Expression , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Neurosecretory Systems/metabolism , Up-Regulation
16.
Eur J Cell Biol ; 96(5): 469-475, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28347547

ABSTRACT

Breast cancer is the most frequent and deadly malignancy in women worldwide. Despite national screening programs combined with new treatments relapse rate remain high and new therapies are needed. From previous work, we identified PACE4, a member of the proprotein convertase (PCs) family of endoproteases, as a novel therapeutic target in prostate cancer. In the present study we asked the question if PACE4 could also be a potential target in breast cancer. In clinical samples of breast adenocarcinoma, we observed a specific overexpression of PACE4 in the estrogen-receptor (ER) positive subtype. We therefore looked for a breast cancer cell line model which would be representative and thus focused on the ZR-75-1 since it both expresses PACE4 and is estrogen-receptor positive. We compared stable knockdowns of furin, PACE4 and PC7 in the estrogen-receptor-positive cell line ZR-75-1 to evaluate their respective contribution to cell growth and tumor progression. PACE4 was the only PC displaying an impact on cell growth. A PACE4 peptide-based inhibitor (C23) was tested and shown to decrease proliferation of ZR-75-1 cells in cell based assays. C23 also had potent effects of tumor progression in vivo on xenografts of the ZR-75-1 cell line in athymic nude mice. Thus, PACE4-silencing and systemic administration of a PACE4 inhibitor resulted in hindered tumor progression with reduction in proliferative indices and increased cell quiescence assessed with biomarkers. Our results suggest that PACE4 is a promising target for estrogen-receptor-positive breast cancer.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , Heterografts , Humans , Mice , Mice, Nude , Receptors, Estrogen/biosynthesis
17.
Eur J Cell Biol ; 96(5): 476-485, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28483279

ABSTRACT

PACE4, one of the seven members of the proprotein convertase family, plays an important role in the progression of prostate cancer. Therefore, its inhibition has become an attractive target to develop new therapies against this disease. Recently, we have developed a highly potent and selective PACE4 inhibitor, known as the multi-Leu peptide with the following sequence Ac-LLLLRVKR-NH2. Herein, with the aim of improving the stability profile of this inhibitor for potential in vivo application, we investigated the impact of different cyclization strategies. The inhibitory activity of new peptides was tested and compared to their linear counterparts. The potent analogues were further selected for stability evaluation. Our results showed that the cyclization involving a C-terminal carboxylic acid (head-to-tail or side chain-to-tail) led to compounds with significantly diminished inhibitory potency towards PACE4, indicating that an appropriate balance between rigidity and flexibility of the structure is necessary to allow the optimal binding with the enzyme. On the other hand, the modification within a multi-Leu core in combination with the incorporation of a C-terminal 4-amidinobenzylamide (Amba) residue yielded potent cyclic analogues. The best compound derived from this group, (&)[Mpa]LLLC(&)RVK[Amba] (where & indicates cyclization, Mpa - 3-mercaptopropionic acid), exhibited promising overall profile comprising of potent inhibitory effect against PACE4 and prostate cancer cell lines as well as improved stability. We believe that this cyclic framework could be further used to design even more potent and stable PACE4 inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Proprotein Convertases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Stability , Enzyme Inhibitors/pharmacology , Humans , Male , Prostatic Neoplasms , Serine Endopeptidases
18.
Cancer Res ; 77(24): 6863-6879, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28993410

ABSTRACT

Inhibition of PACE4, a proprotein convertase that is overexpressed in prostate cancer, has been shown to block cancer progression in an androgen-independent manner. However, the basis for its overexpression and its growth-inhibitory effects are mitigated and uncertain. Here, we report that PACE4 pre-mRNA undergoes DNA methylation-sensitive alternative splicing of its terminal exon 3' untranslated region, generating an oncogenic, C-terminally modified isoform (PACE4-altCT). We found this isoform to be strongly expressed in prostate cancer cells, where it displayed an enhanced autoactivating process and a distinct intracellular routing that prevented its extracellular secretion. Together, these events led to a dramatic increase in processing of the progrowth differentiation factor pro-GDF15 as the first PACE4 substrate to be identified in prostate cancer. We detected robust expression of PACE4-altCT in other cancer types, suggesting that an oncogenic switch for this proenzyme may offer a therapeutic target not only in advanced prostate cancer but perhaps also more broadly in human cancer. Cancer Res; 77(24); 6863-79. ©2017 AACR.


Subject(s)
Alternative Splicing/genetics , Genes, Switch , Neoplasms/genetics , Oncogenes , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , DNA Methylation , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Neoplasms/pathology , Oncogenes/genetics , Proprotein Convertases/metabolism , Prostatic Neoplasms/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Serine Endopeptidases/metabolism
19.
ChemMedChem ; 12(15): 1169-1172, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28722823

ABSTRACT

PACE4, a member of the proprotein convertases (PCs) family of serine proteases, is a validated target for prostate cancer. Our group has developed a potent and selective PACE4 inhibitor: Ac-LLLLRVKR-NH2 . In seeking for modifications to increase the selectivity of this ligand toward PACE4, we replaced one of its P3 Val methyl groups with a basic group capable of forming a salt bridge with D160 of PACE4. The resulting inhibitor is eight times more potent than the P3 Val parent inhibitor and two times more selective over furin, because the equivalent salt bridge with furin E257 is not optimal. Moreover, the ß-branched nature of the new P3 residue favors the extended ß-sheet conformation usually associated with substrates of proteases. This work provides new insight for better understanding of ß-sheet backbone-backbone interactions between serine proteases and their peptidic ligands.


Subject(s)
Drug Design , Peptides/pharmacology , Proprotein Convertases/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Proprotein Convertases/metabolism , Salts/chemical synthesis , Salts/chemistry , Salts/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
20.
J Med Chem ; 60(7): 2732-2744, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28287731

ABSTRACT

The proprotein convertase family of enzymes includes seven endoproteases with significant redundancy in their cleavage activity. We previously described the peptide Ac-LLLLRVK-Amba that displays potent inhibitory effects on both PACE4 and prostate cancer cell lines proliferation. Herein, the molecular determinants for PACE4 and furin inhibition were investigated by positional scanning using peptide libraries that substituted its leucine core with each natural amino acid. We determined that the incorporation of basic amino acids led to analogues with improved inhibitory potency toward both enzymes, whereas negatively charged residues significantly reduced it. All the remaining amino acids were in general well tolerated, with the exemption of the P6 position. However, not all of the potent PACE4 inhibitors displayed antiproliferative activity. The best analogues were obtained by the incorporation of the Ile residue at the P5 and P6 positions. These substitutions led to inhibitors with increased PACE4 selectivity and potent antiproliferative effects.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Furin/antagonists & inhibitors , Peptides/chemistry , Peptides/pharmacology , Proprotein Convertases/antagonists & inhibitors , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Furin/metabolism , Humans , Male , Peptide Library , Proprotein Convertases/metabolism , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Serine Endopeptidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL