Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Life Sci Alliance ; 5(12)2022 07 29.
Article in English | MEDLINE | ID: mdl-35905995

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) and its cognate receptor MET play several essential roles in embryogenesis and regeneration in postnatal life of epithelial organs such as the liver, kidney, lung, and pancreas, prompting a strong interest in harnessing HGF/SF-MET signalling for regeneration of epithelial organs after acute or chronic damage. The limited stability and tissue diffusion of native HGF/SF, however, which reflect the tightly controlled, local mechanism of action of the morphogen, have led to a major search of HGF/SF mimics for therapy. In this work, we describe the rational design, production, and characterization of K1K1, a novel minimal MET agonist consisting of two copies of the kringle 1 domain of HGF/SF in tandem orientation. K1K1 is highly stable and displays biological activities equivalent or superior to native HGF/SF in a variety of in vitro assay systems and in a mouse model of liver disease. These data suggest that this engineered ligand may find wide applications in acute and chronic diseases of the liver and other epithelial organs dependent of MET activation.


Subject(s)
Hepatocyte Growth Factor , Kringles , Animals , Dimerization , Hepatocyte Growth Factor/metabolism , Liver/metabolism , Mice , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/metabolism
2.
Angew Chem Int Ed Engl ; 60(42): 22745-22752, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34142433

ABSTRACT

Designing synthetic surrogates of functional proteins is an important, albeit challenging, task in the field of chemistry. A strategy toward the design of synthetic agonists for growth factor or cytokine receptors that elicit a desired signal activity has been in high demand, as such ligands hold great promise as safer and more effective therapeutics. In the present study, we used a DNA aptamer as a building block and described the strategy-guided design of a synthetic receptor agonist with fine-tuned agonism. The developed synthetic partial agonist can regulate therapeutically relevant cellular activities by eliciting fine-tuned receptor signaling.


Subject(s)
Aptamers, Nucleotide/metabolism , Intercellular Signaling Peptides and Proteins/agonists , Receptors, Cytokine/agonists , A549 Cells , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Cell Movement/drug effects , Dimerization , Hepatocyte Growth Factor/agonists , Hepatocyte Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Microscopy, Fluorescence , Protein Binding , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Receptors, Cytokine/metabolism , Signal Transduction/drug effects
3.
Drug Dev Res ; 82(1): 123-132, 2021 02.
Article in English | MEDLINE | ID: mdl-32830369

ABSTRACT

Hepatocyte growth factor (HGF) is central to liver regeneration. The Internalin B (InlB) protein is a virulence factor produced by the pathogenic bacterium Listeria monocytogenes. InlB is known to mimic HGF activity by interacting with the HGF receptor (HGFR) and activating HGFR-controlled signaling pathways. We expressed and purified the HGFR-binding InlB domain, InlB321/15, cloned from the fully virulent clinical L. monocytogenes strain. HGFR and Erk1/2 phosphorylation was determined using Western blotting. The capacity of InlB321/15 to bind HGFR was measured using microscale thermophoresis. Liver regeneration was studied in a model of 70% partial hepatectomy (70%PHx) in male Wistar rats. The nuclear grade parameters were quantified using manual (percentage of binuclear hepatocytes), automated (nuclear diameters), or combined (Ki67 proliferation index) scoring methods. Purified InlB321/15 stimulated HGFR and Erk1/2 phosphorylation and accelerated the proliferation of HepG2 cells. InlB321/15 bound HGFR with Kd = 7.4 ± 1.3 nM. InlB321/15 injected intravenously on the second, fourth, and sixth days after surgery recovered the liver mass and improved the nuclear grade parameters. Seven days post 70% PHx, the liver weight indexes were 2.9 and 2.0%, the hepatocyte proliferation indexes were 19.8 and 0.6%, and the percentages of binucleated hepatocytes were 6.7 and 4.0%, in the InlB321/15-treated and control animals, respectively. Obtained data demonstrated that InlB321/15 improved hepatocyte proliferation and stimulated liver regeneration in animals with 70% hepatectomy.


Subject(s)
Bacterial Proteins/pharmacology , Liver Regeneration/drug effects , Proto-Oncogene Proteins c-met/agonists , Animals , Bacterial Proteins/genetics , Cell Proliferation/drug effects , Hep G2 Cells , Hepatectomy , Humans , Listeria monocytogenes , Male , Proto-Oncogene Proteins c-met/genetics , Rats, Wistar , Recombinant Proteins/pharmacology
4.
Int J Mol Sci ; 21(21)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121208

ABSTRACT

Using a random non-standard peptide integrated discovery system, we obtained cyclic peptides that bind to hepatocyte growth factor (HGF) or mesenchymal-epithelial transition factor. (MET) HGF-inhibitory peptide-8 (HiP-8) selectively bound to two-chain active HGF, but not to single-chain precursor HGF. HGF showed a dynamic change in its molecular shape in atomic force microscopy, but HiP-8 inhibited dynamic change in the molecular shape into a static status. The inhibition of the molecular dynamics of HGF by HiP-8 was associated with the loss of the ability to bind MET. HiP-8 could selectively detect active HGF in cancer tissues, and active HGF probed by HiP-8 showed co-localization with activated MET. Using HiP-8, cancer tissues with active HGF could be detected by positron emission tomography. HiP-8 seems to be applicable for the diagnosis and treatment of cancers. In contrast, based on the receptor dimerization as an essential process for activation, the cross-linking of the cyclic peptides that bind to the extracellular region of MET successfully generated an artificial ligand to MET. The synthetic MET agonists activated MET and exhibited biological activities which were indistinguishable from the effects of HGF. MET agonists composed of cyclic peptides can be manufactured by chemical synthesis but not recombinant protein expression, and thus are expected to be new biologics that are applicable to therapeutics and regenerative medicine.


Subject(s)
Biological Products/pharmacology , Hepatocyte Growth Factor/metabolism , Neoplasms/metabolism , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Binding Sites , Biological Products/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/antagonists & inhibitors , Humans , Neoplasms/drug therapy , Peptides, Cyclic/therapeutic use , Protein Binding , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/chemistry , Signal Transduction/drug effects
5.
Sci Rep ; 9(1): 13495, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530851

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor, cMet, activate biological pathways necessary for repair and regeneration following kidney injury. Because HGF is a highly unstable molecule in its biologically active form, we asked whether a monoclonal antibody (Ab) that displays full agonist activity at the receptor could protect the kidney from fibrosis. We attempted to determine whether the cMet agonistic Ab might reduce fibrosis, the final common pathway for chronic kidney diseases (CKD). A mouse model of kidney fibrosis disease induced by unilateral ureteral obstruction was introduced and subsequently validated with primary cultured human proximal tubular epithelial cells (PTECs). In kidney biopsy specimens from patients with CKD, cMet immunohistochemistry staining showed a remarkable increase compared with patients with normal renal functions. cMet Ab treatment significantly increased the levels of phospho-cMet and abrogated the protein expression of fibrosis markers such as fibronectin, collagen 1, and αSMA as well as Bax2, which is a marker of apoptosis triggered by recombinant TGF-ß1 in PTECs. Remarkably, injections of cMet Ab significantly prevented kidney fibrosis in obstructed kidneys as quantified by Masson trichrome staining. Consistent with these data, cMet Ab treatment decreased the expression of fibrosis markers, such as collagen1 and αSMA, whereas the expression of E-cadherin, which is a cell-cell adhesion molecule, was restored. In conclusion, cMet-mediated signaling may play a considerable role in kidney fibrosis. Additionally, the cMet agonistic Ab may be a valuable substitute for HGF because it is more easily available in a biologically active, stable, and purified form.


Subject(s)
Antibodies, Monoclonal/pharmacology , Protective Agents/pharmacology , Proto-Oncogene Proteins c-met/agonists , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Animals , Biomarkers , Disease Models, Animal , Endothelial Cells/metabolism , Fibrosis , Gene Expression , Humans , Immunohistochemistry , Kidney Tubules, Proximal/metabolism , Mice , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology
6.
Sci Rep ; 8(1): 16492, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405161

ABSTRACT

Non-native ligands for growth factor receptors that are generated by chemical synthesis are applicable to therapeutics. However, non-native ligands often regulate cellular signaling and biological responses in a different manner than native ligands. Generation of surrogate ligands comparable to native ligands is a challenging need. Here we investigated changes in signal transduction and gene expression evoked by a bivalent macrocyclic peptide (aMD5-PEG11) capable of high-affinity binding to the MET/hepatocyte growth factor (HGF) receptor. Binding of aMD5-PEG11 to the MET extracellular region was abolished by deletion of the IPT3-IPT4 domain, indicating the involvement of IPT3-IPT4 in the binding of aMD5-PEG11 to the MET receptor. aMD5-PEG11 induced dimerization and activation of the MET receptor and promoted cell migration that was comparable to induction of these activities by HGF. Signal activation profiles indicated that aMD5-PEG11 induced phosphorylation of intracellular signaling molecules, with a similar intensity and time dependency as HGF. In 3-D culture, aMD5-PEG11 as well as HGF induced epithelial tubulogenesis and up-regulated the same sets of functionally classified genes involved in multicellular organism development. Thus, a non-native surrogate ligand that consisted of a bivalent macrocyclic peptide can serve as an artificial MET receptor agonist that functionally substitutes for the native ligand, HGF.


Subject(s)
Gene Expression Regulation/drug effects , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-met/agonists , Receptors, Artificial/agonists , Signal Transduction/drug effects , Transcriptome , Cell Line , Computational Biology/methods , Gene Expression Profiling , Molecular Structure , Peptides, Cyclic/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Proto-Oncogene Proteins c-met/chemistry , Receptors, Artificial/chemistry
7.
Arch Dermatol Res ; 310(10): 849-856, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30306262

ABSTRACT

Studies of factors affecting wound-healing rates are encouraged by a critical need for new treatments to manage an increasing burden of non-healing wounds. The InlB protein produced by the Gram-positive bacterium Listeria monocytogenes is an agonist of the tyrosine kinase receptor c-Met and a functional analog of the hepatocyte growth factor (HGF), which is a mammalian ligand of c-Met. The recombinant InlB321 protein, which is the c-Met-binding InlB domain (amino acids 31-321), was cloned from the L. monocytogenes serovar 4b clinical strain VIMHA015 and serovar 1/2a strain EGDe (InlB321/15 and InlB321/EGDe, respectively). Both InlB321 variants stimulated proliferation of endothelial HUVEC cells. InlB321/15 was more active in Erk1/2 phosphorylation assay, and more potent than InlB321/EGDe in the 2D-scratch wound-healing assay. Scratch closure reached 86%, 29% and 72% for InlB321/15, InlB321/EGDe and HGF, respectively, 72 h post-wounding (p < 0.05). Topically applied glycerol-mixed InlB321/15 (300 µg ml- 1) increased abrasion wound-healing rates in mice. The 50% wound closing time (CT50) was reduced by InlB321/15 (4.18 ± 0.91 days; CI: 3.05; 5.31) compared with control animals (5.51 ± 1.21 days; CI: 4.01; 7.01; p < 0.05). Taken together, obtained results suggested a potential of InlB321/15 as a means of accelerating wound healing.


Subject(s)
Bacterial Proteins/pharmacology , Hepatocyte Growth Factor/metabolism , Membrane Proteins/pharmacology , Skin/injuries , Wound Healing/drug effects , Animals , Bacterial Proteins/adverse effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Listeria monocytogenes/metabolism , Membrane Proteins/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/agonists , Recombinant Proteins/pharmacology
8.
Int J Mol Sci ; 19(10)2018 Oct 12.
Article in English | MEDLINE | ID: mdl-30322054

ABSTRACT

Non-native ligands for growth factor receptors with distinct chemical properties and different biological activities have the potential to become therapeutic applications. We previously generated MET/hepatocyte growth factor (HGF) receptor agonists using bivalent macrocyclic peptides. The highest MET-activating agonists exhibited biological activity that was indistinguishable from the effects of HGF. In this study, we investigated MET activation, signal characteristics, and biological responses induced by a macrocyclic peptide partial agonist known as aML5-PEG11. aML5-PEG11 induced weak tyrosine phosphorylation of MET while enhancing cell migration with potency comparable to HGF. aML5-PEG11 induced marked AKT (protein kinase B) and ERK (extracellular signal-regulated kinase) activation at a comparable potency and time-dependency to HGF, which suggests that enhancement of cell motility is attributable to activation of these molecules. In a 3-D culture of bile duct cancer cells in collagen gel, HGF induced robust activation of MET, ERK, and AKT, which was associated with enhanced expression of genes involved in bile duct development and subsequent branching of tubulogenesis. In contrast, aML5-PEG11 induced marginal activation of MET, ERK, and AKT (levels near the detection limits), which was associated with failure to enhance the expression of genes involved in bile duct development and a lack of tubulogenic response. Thus, MET activation by aML5-PEG11 couples to biological responses differently from HGF in an extracellular context-dependent manner.


Subject(s)
Hepatocyte Growth Factor/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Transcriptional Activation , Cell Line, Tumor , Cell Movement/drug effects , Humans , MAP Kinase Signaling System/drug effects , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Peptides/chemistry , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-met/agonists
9.
Curr Pharm Biotechnol ; 18(4): 309-317, 2017.
Article in English | MEDLINE | ID: mdl-28155605

ABSTRACT

BACKGROUND: Myogenic progenitor cells (activated satellite cells) are able to express both HGF and its receptor cMet. After muscle injury, HGF-Met stimulation promotes activation and primary division of satellite cells. MAGIC-F1 (Met-Activating Genetically Improved Chimeric Factor-1) is an engineered protein that contains two human Met-binding domains that promotes muscle hypertrophy. MAGIC-F1 protects myogenic precursors against apoptosis and increases their fusion ability enhancing muscle differentiation. Hemizygous and homozygous Magic-F1 transgenic mice displayed constitutive muscle hypertrophy. METHODS: Here we describe microarray analysis on Magic-F1 myogenic progenitor cells showing an altered gene signatures on muscular hypertrophy and angiogenesis compared to wild-type cells. In addition, we performed a functional analysis on Magic-F1+/+ transgenic mice versus controls using treadmill test. RESULTS: We demonstrated that Magic-F1+/+ mice display an increase in muscle mass and cross-sectional area leading to an improvement in running performance. Moreover, the presence of MAGIC-F1 affected positively the vascular network, increasing the vessel number in fast twitch fibers. Finally, the gene expression profile analysis of Magic-F1+/+ satellite cells evidenced transcriptomic changes in genes involved in the control of muscle growth, development and vascularisation. CONCLUSION: We showed that MAGIC-F1-induced muscle hypertrophy affects positively vascular network, increasing vessel number in fast twitch fibers. This was due to unique features of mammalian skeletal muscle and its remarkable ability to adapt promptly to different physiological demands by modulating the gene expression profile in myogenic progenitors.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-met/agonists , Recombinant Proteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cells, Cultured , Exercise Test , Female , Gene Expression , Humans , Hypertrophy , Mice , Mice, Transgenic , Muscle Development/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/genetics , Recombinant Proteins/genetics
10.
Biomed Res Int ; 2015: 192406, 2015.
Article in English | MEDLINE | ID: mdl-26448928

ABSTRACT

AIMS: Inhibitors of the MET pathway hold promise in the treatment for metastatic kidney cancer. Assessment of predictive biomarkers may be necessary for appropriate patient selection. Understanding MET expression in metastases and the correlation to the primary site is important, as distant tissue is not always available. METHODS AND RESULTS: MET immunofluorescence was performed using automated quantitative analysis and a tissue microarray containing matched nephrectomy and distant metastatic sites from 34 patients with clear cell renal cell carcinoma. Correlations between MET expressions in matched primary and metastatic sites and the extent of heterogeneity were calculated. The mean expression of MET was not significantly different between primary tumors when compared to metastases (P = 0.1). MET expression weakly correlated between primary and matched metastatic sites (R = 0.5) and a number of cases exhibited very high levels of discordance between these tumors. Heterogeneity within nephrectomy specimens compared to the paired metastatic tissues was not significantly different (P = 0.39). CONCLUSIONS: We found that MET expression is not significantly different in primary tumors than metastatic sites and only weakly correlates between matched sites. Moderate concordance of MET expression and significant expression heterogeneity may be a barrier to the development of predictive biomarkers using MET targeting agents.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Female , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Male , Neoplasm Metastasis , Nephrectomy , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/biosynthesis , Retrospective Studies , Tissue Array Analysis
11.
Stem Cell Reports ; 4(5): 939-52, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25937370

ABSTRACT

The differentiation of pluripotent stem cells to hepatocytes is well established, yet current methods suffer from several drawbacks. These include a lack of definition and reproducibility, which in part stems from continued reliance on recombinant growth factors. This has remained a stumbling block for the translation of the technology into industry and the clinic for reasons associated with cost and quality. We have devised a growth-factor-free protocol that relies on small molecules to differentiate human pluripotent stem cells toward a hepatic phenotype. The procedure can efficiently direct both human embryonic stem cells and induced pluripotent stem cells to hepatocyte-like cells. The final population of cells demonstrates marker expression at the transcriptional and protein levels, as well as key hepatic functions such as serum protein production, glycogen storage, and cytochrome P450 activity.


Subject(s)
Cell Differentiation/drug effects , Hepatocytes/cytology , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Blood Proteins/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Dimethyl Sulfoxide/pharmacology , Glycogen/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/metabolism , Humans , Microscopy, Fluorescence , Oligopeptides/pharmacology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/metabolism
12.
Nat Commun ; 6: 6373, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25758345

ABSTRACT

Hepatocyte growth factor (HGF) receptor, also known as Met, is a member of the receptor tyrosine kinase family. The Met-HGF interaction regulates various signalling pathways involving downstream kinases, such as Akt and Erk. Met activation is implicated in wound healing of tissues via multiple biological responses triggered by the above-mentioned signalling cascade. Here we report the development of artificial Met-activating dimeric macrocycles. We identify Met-binding monomeric macrocyclic peptides by means of the RaPID (random non-standard peptide integrated discovery) system, and dimerize the respective monomers through rational design. These dimeric macrocycles specifically and strongly activate Met signalling pathways through receptor dimerization and induce various HGF-like cellular responses, such as branching morphogenesis, in human cells. This work suggests our approach for generating dimeric macrocycles as non-protein ligands for cell surface receptors can be useful for developing potential therapeutics with a broad range of potential applications.


Subject(s)
Antineoplastic Agents/pharmacology , Hepatocyte Growth Factor/metabolism , Morphogenesis/drug effects , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-met/agonists , Antineoplastic Agents/chemical synthesis , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Dimerization , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Hepatocyte Growth Factor/genetics , Humans , Ligands , Morphogenesis/genetics , Peptides, Cyclic/chemical synthesis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Wound Healing/drug effects
13.
Immunotherapy ; 6(12): 1243-53, 2014.
Article in English | MEDLINE | ID: mdl-25524381

ABSTRACT

In 2014, outcomes for patients with advanced gastric cancer remain extremely poor with a high level of unmet need regarding effective therapeutic options. However, recent years have seen increasing interest in the role of the MET signaling pathway in this disease subtype, leading to the development and evaluation of MET-targeted therapeutics. Rilotumumab is a monoclonal antibody directed against hepatocyte growth factor, the only known ligand for the MET receptor. It is an unlicensed product which is currently undergoing evaluation in a randomized Phase III trial in 'MET-positive' gastric cancer. Here we discuss the background to the treatment of gastric cancer as well as the characteristics of rilotumumab and reported results with this agent in the trials performed to date.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hepatocyte Growth Factor/metabolism , Immunotherapy/methods , Proto-Oncogene Proteins c-met/agonists , Stomach Neoplasms/therapy , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Hepatocyte Growth Factor/immunology , Humans , Molecular Targeted Therapy , Proto-Oncogene Proteins c-met/metabolism , Randomized Controlled Trials as Topic , Signal Transduction/drug effects , Stomach Neoplasms/immunology
14.
FEBS Lett ; 588(24): 4831-7, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25451235

ABSTRACT

Hepatocyte growth factor (HGF), through activation of the c-MET receptor, mediates biological processes critical for tissue regeneration; however, its clinical application is limited by protein instability and poor recombinant expression. We previously engineered an HGF fragment (eNK1) that possesses increased stability and expression yield and developed a c-MET agonist by coupling eNK1 through an introduced cysteine residue. Here, we further characterize this eNK1 dimer and show it elicits significantly greater c-MET activation, cell migration, and proliferation than the eNK1 monomer. The efficacy of the eNK1 dimer was similar to HGF, suggesting its promise as a c-MET agonist.


Subject(s)
Hepatocyte Growth Factor/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Engineering , Protein Multimerization , Proto-Oncogene Proteins c-met/agonists , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Stability , Protein Structure, Quaternary , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Temperature
15.
PLoS One ; 9(8): e105278, 2014.
Article in English | MEDLINE | ID: mdl-25148256

ABSTRACT

c-Met, the receptor for Hepatocyte Growth Factor (HGF), overexpressed and deregulated in Hepatocellular Carcinoma (HCC). Caveolin 1 (CAV1), a plasma membrane protein that modulates signal transduction molecules, is also overexpressed in HCC. The aim of this study was to investigate biological and clinical significance of co-expression and activation of c-Met and CAV1 in HCC. We showed that c-Met and CAV1 were co-localized in HCC cells and HGF treatment increased this association. HGF-triggered c-Met activation caused a concurrent rise in both phosphorylation and expression of CAV1. Ectopic expression of CAV1 accelerated c-Met signaling, resulted in enhanced migration, invasion, and branching-morphogenesis. Silencing of CAV1 downregulated c-Met signaling, and decreased migratory/invasive capability of cells and attenuated branching morphogenesis. In addition, activation and co-localization of c-Met and CAV1 were elevated during hepatocarcinogenesis. In conclusion reciprocal activating crosstalk between c-Met and CAV1 promoted oncogenic signaling of c-Met contributed to the initiation and progression of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caveolin 1/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Hepatocyte Growth Factor/pharmacology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Neoplasm Invasiveness , Phosphorylation , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA Interference , Signal Transduction
16.
Cell Death Dis ; 5: e1185, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24743740

ABSTRACT

Met, the tyrosine kinase receptor for hepatocyte growth factor (HGF), mainly activates prosurvival pathways, including protection from apoptosis. In this work, we investigated the cardioprotective mechanisms of Met activation by agonist monoclonal antibodies (mAbs). Cobalt chloride (CoCl2), a chemical mimetic of hypoxia, was used to induce cardiac damage in H9c2 cardiomyoblasts, which resulted in reduction of cell viability by (i) caspase-dependent apoptosis and (ii) - surprisingly - autophagy. Blocking either apoptosis with the caspase inhibitor benzyloxycarbonyl-VAD-fluoromethylketone or autophagosome formation with 3-methyladenine prevented loss of cell viability, which suggests that both processes contribute to cardiomyoblast injury. Concomitant treatment with Met-activating antibodies or HGF prevented apoptosis and autophagy. Pro-autophagic Redd1, Bnip3 and phospho-AMPK proteins, which are known to promote autophagy through inactivation of the mTOR pathway, were induced by CoCl2. Mechanistically, Met agonist antibodies or HGF prevented the inhibition of mTOR and reduced the flux of autophagosome formation. Accordingly, their anti-autophagic function was completely blunted by Temsirolimus, a specific mTOR inhibitor. Targeted Met activation was successful also in the setting of low oxygen conditions, in which Met agonist antibodies or HGF demonstrated anti-apoptotic and anti-autophagic effects. Activation of the Met pathway is thus a promising novel therapeutic tool for ischaemic injury.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cardiotonic Agents/pharmacology , Cytoprotection/drug effects , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-met/agonists , Animals , Cell Line , Cobalt , Hepatocyte Growth Factor/pharmacology , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
17.
Proc Natl Acad Sci U S A ; 108(32): 13035-40, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788476

ABSTRACT

The Met receptor tyrosine kinase and its ligand hepatocyte growth factor (HGF) play an important role in mediating both tumor progression and tissue regeneration. The N-terminal and first Kringle domains (NK1) of HGF comprise a naturally occurring splice variant that retains the ability to activate the Met receptor. However, NK1 is a weak agonist and is relatively unstable, limiting its therapeutic potential. Here, we engineered NK1 mutants with improved biochemical and biophysical properties that function as Met receptor agonists or antagonists. We first engineered NK1 for increased stability and recombinant expression yield using directed evolution. The NK1 variants isolated from our library screens acted as weak Met receptor antagonists due to a mutation at the NK1 homodimerization interface. We introduced point mutations that restored this NK1 homodimerization interface to create an agonistic ligand, or that further disrupted this interface to create more effective antagonists. The rationally engineered antagonists exhibited melting temperatures up to approximately 64 °C, a 15 °C improvement over antagonists derived from wild-type NK1, and approximately 40-fold improvement in expression yield. Next, we created disulfide-linked NK1 homodimers through introduction of an N-terminal cysteine residue. These covalent dimers exhibited nearly an order of magnitude improved agonistic activity compared to wild-type NK1, approaching the activity of full-length HGF. Moreover, covalent NK1 dimers formed from agonistic or antagonistic monomeric subunits elicited similar activity, further signifying that NK1 dimerization mediates agonistic activity. These engineered NK1 proteins are promising candidates for therapeutic development and will be useful tools for further exploring determinants of Met receptor activation.


Subject(s)
Hepatocyte Growth Factor/metabolism , Peptide Fragments/metabolism , Protein Engineering/methods , Proto-Oncogene Proteins c-met/agonists , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line , Dogs , Hepatocyte Growth Factor/chemistry , Humans , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/chemistry , Peptide Library , Point Mutation/genetics , Protein Binding , Protein Multimerization , Protein Stability
18.
Chemistry ; 17(4): 1319-26, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21243700

ABSTRACT

Variation of the cultivation conditions for Aspergillus glaucus led to the discovery of two novel spirocyclic aromatic polyketides, aspergiolides C (3) and D (4). Their constitutions were elucidated by a combination of spectroscopic methods and isotope-labeling experiments. Aspergiolides C (3) and D (4) occur as racemic mixtures, the resolution of which was succeeded by HPLC on a chiral phase. The absolute configurations of their enantiomers were assigned online, from the peaks in the chromatogram, by a combination of HPLC-CD and quantum chemical CD calculations. Both compounds were found to inhibit the kinase activities of the receptor tyrosine kinases (RTKs) c-Met, Ron, and c-Src with low-micromolar IC(50)s. The enantiomers of 3 were resolved by HPLC on a chiral phase. Both enantiomers showed a comparable inhibition of the HGF-induced autophosphorylation of c-Met and of subsequent cell migration.


Subject(s)
Anthraquinones/chemistry , Anthraquinones/pharmacology , Aspergillus/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Hepatocyte Growth Factor/chemistry , Macrolides/chemistry , Macrolides/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/agonists , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Anthraquinones/isolation & purification , Aspergillus/chemistry , Cell Movement/drug effects , Hepatocyte Growth Factor/metabolism , Humans , Macrolides/isolation & purification , Molecular Structure , Phosphorylation , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/metabolism , Quantum Theory , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...