27 results on '"Lerner EC"'
Search Results
2. CD8 + T cells maintain killing of MHC-I-negative tumor cells through the NKG2D-NKG2DL axis.
- Author
-
Lerner EC, Woroniecka KI, D'Anniballe VM, Wilkinson DS, Mohan AA, Lorrey SJ, Waibl-Polania J, Wachsmuth LP, Miggelbrink AM, Jackson JD, Cui X, Raj JA, Tomaszewski WH, Cook SL, Sampson JH, Patel AP, Khasraw M, Gunn MD, and Fecci PE
- Subjects
- Animals, Humans, Mice, Antigens metabolism, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I metabolism, NK Cell Lectin-Like Receptor Subfamily K genetics, NK Cell Lectin-Like Receptor Subfamily K metabolism, CD8-Positive T-Lymphocytes pathology, Neoplasms genetics
- Abstract
The accepted paradigm for both cellular and anti-tumor immunity relies upon tumor cell killing by CD8
+ T cells recognizing cognate antigens presented in the context of target cell major histocompatibility complex (MHC) class I (MHC-I) molecules. Likewise, a classically described mechanism of tumor immune escape is tumor MHC-I downregulation. Here, we report that CD8+ T cells maintain the capacity to kill tumor cells that are entirely devoid of MHC-I expression. This capacity proves to be dependent instead on interactions between T cell natural killer group 2D (NKG2D) and tumor NKG2D ligands (NKG2DLs), the latter of which are highly expressed on MHC-loss variants. Necessarily, tumor cell killing in these instances is antigen independent, although prior T cell antigen-specific activation is required and can be furnished by myeloid cells or even neighboring MHC-replete tumor cells. In this manner, adaptive priming can beget innate killing. These mechanisms are active in vivo in mice as well as in vitro in human tumor systems and are obviated by NKG2D knockout or blockade. These studies challenge the long-advanced notion that downregulation of MHC-I is a viable means of tumor immune escape and instead identify the NKG2D-NKG2DL axis as a therapeutic target for enhancing T cell-dependent anti-tumor immunity against MHC-loss variants., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
3. Risk of Tract Seeding Following Laser Interstitial Thermal Therapy for Brain Tumors.
- Author
-
Haskell-Mendoza AP, Srinivasan ES, Lerner EC, Edwards RM, Schwalb AM, Jackson JD, Hardigan AA, Vaios EJ, and Fecci PE
- Subjects
- Humans, Retrospective Studies, Progression-Free Survival, Lasers, Brain Neoplasms pathology, Laser Therapy methods
- Abstract
Background: The management of intracranial oncological disease remains a significant challenge despite advances in systemic cancer therapy. Laser interstitial thermal therapy (LITT) represents a novel treatment for local control of brain tumors through photocoagulation with a stereotactically implanted laser fiber. Because the use of laser interstitial thermal therapy continues to increase within neurosurgery, characterization of LITT is necessary to improve outcomes., Objective: To quantify the risk of tumor seeding along the laser fiber tract in patients receiving LITT for primary or metastatic brain tumors at a high-volume treatment center., Methods: We retrospectively reviewed all patients receiving LITT from 2015 to 2021 at our medical center. Patients with biopsy-confirmed tumors were included in this study. Tract seeding was identified as discontinuous, newly enhancing tumor along the LITT tract., Results: Fifty-six patients received LITT for biopsy-confirmed tumors from 2015 to 2021, with tract seeding identified in 3 (5.4%). Twenty-nine (51.8%) patients had gliomas, while the remainder had metastases, of which lung was the most common histology (20 patients, 74%). Tract seeding was associated with ablation proceeding inward from superficial tumor margin closest to the cranial entry point ( P = .03). Patients with tract seeding had a shorter median time to progression of 1.1 (0.1-1.3) months vs 4.2 (2.2-8.6) months ( P = .03)., Conclusion: Although the risk of tract seeding after LITT is reassuringly low, it is associated with decreased progression-free survival. This risk may be related to surgical technique or experience. Follow-up radiosurgery to the LITT tract has the potential to prevent this complication., (Copyright © Congress of Neurological Surgeons 2023. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
4. Erratum to "E.C. Lerner, E.S. Srinivasan, G. Broadwater, et al. Factors Associated With New-Onset Seizures Following Stereotactic Radiosurgery for Newly Diagnosed Brain Metastases. Adv Radiat Oncol. 2022;7:101054."
- Author
-
Lerner EC, Srinivasan ES, Broadwater G, Haskell-Mendoza AP, Vaios EJ, Edwards RM, Wachsmuth LP, Huie D, Floyd SR, Adamson JD, and Fecci PE
- Abstract
[This corrects the article DOI: 10.1016/j.adro.2022.101054.]., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
5. Factors Associated With New-Onset Seizures Following Stereotactic Radiosurgery for Newly Diagnosed Brain Metastases.
- Author
-
Lerner EC, Srinivasan ES, Broadwater G, Haskell-Mendoza AP, Edwards RM, Huie D, Vaios EJ, Floyd SR, Adamson JD, and Fecci PE
- Abstract
Purpose: Stereotactic radiosurgery (SRS) is a highly effective therapy for newly diagnosed brain metastases. Prophylactic antiepileptic drugs are no longer routinely used in current SRS practice, owing to a perceived low overall frequency of new-onset seizures and potential side effects of medications. It is nonetheless desirable to prevent unwanted side effects following SRS. Risk factors for new-onset seizures after SRS have not been well established. As such, we aimed to characterize variables associated with increased seizure risk., Methods and Materials: Patients treated with SRS for newly diagnosed brain metastases between 2013 and 2016 were retrospectively reviewed at a single institution. Data on baseline demographics, radiation parameters, and clinical courses were collected., Results: The cohort consisted of 305 patients treated with SRS without prior seizure history. Median age and baseline Karnofsky Performance Scale score were 64 years (interquartile range, 55-70) and 80 (interquartile range, 80-90), respectively. Twenty-six (8.5%) patients developed new-onset seizures within 3 months of SRS. There was no association between new-onset seizures and median baseline Karnofsky Performance Scale score, prior resection, or prior whole brain radiation therapy. There were significant differences in the combined total irradiated volume (12.5 vs 3.7 cm
3 , P < .001), maximum single lesion volume (8.8 vs 2.8 cm3 , P = .003), lesion diameter (3.2 vs 2.0 cm, P = .003), and number of lesions treated (3 vs 1, P = .018) between patients with and without new-onset seizures, respectively. On multivariate logistic regression, total irradiated volume (odds ratio, 1.09 for every 1-cm1 increase in total volume; confidence interval, 1.02-1.17; P = .016) and pre-SRS neurologic symptoms (odds ratio, 3.08; 95% confidence interval, 1.19-7.99; P = .020) were both significantly correlated with odds of seizures following SRS., Conclusions: Our data suggest that larger total treatment volume and the presence of focal neurologic deficits at presentation are associated with new-onset seizures within 3 months of SRS. High-risk patients undergoing SRS may benefit from counseling or prophylactic antiseizure therapy., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
6. Laser ablation: Heating up the anti-tumor response in the intracranial compartment.
- Author
-
Lerner EC, Edwards RM, Wilkinson DS, and Fecci PE
- Subjects
- Blood-Brain Barrier, Heating, Humans, Brain Neoplasms drug therapy, Hyperthermia, Induced, Laser Therapy
- Abstract
Immunotherapies, such as immune checkpoint inhibition (ICI), have had limited success in treating intracranial malignancies. These failures are due partly to the restrictive blood-brain-barrier (BBB), the profound tumor-dependent induction of local and systemic immunosuppression, and immune evasion exhibited by these tumors. Therefore, novel approaches must be explored that aim to overcome these stringent barriers. LITT is an emerging treatment for brain tumors that utilizes thermal ablation to kill tumor cells. LITT provides an additional therapeutic benefit by synergizing with ICI and systemic chemotherapies to strengthen the anti-tumor immune response. This synergistic relationship involves transient disruption of the BBB and local augmentation of immune function, culminating in increased CNS drug penetrance and improved anti-tumor immunity. In this review, we will provide an overview of the challenges facing immunotherapy for brain tumors, and discuss how LITT may synergize with the endogenous anti-tumor response to improve the efficacy of ICI., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
7. Pushing Past the Blockade: Advancements in T Cell-Based Cancer Immunotherapies.
- Author
-
Waibl Polania J, Lerner EC, Wilkinson DS, Hoyt-Miggelbrink A, and Fecci PE
- Subjects
- Animals, Biomarkers, Tumor, Combined Modality Therapy adverse effects, Combined Modality Therapy methods, Disease Management, Disease Susceptibility, Humans, Immunity, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Neoplasms diagnosis, Neoplasms metabolism, Prognosis, T-Lymphocytes metabolism, Treatment Outcome, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Immunotherapy adverse effects, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy, T-Lymphocytes immunology
- Abstract
Successful cancer immunotherapies rely on a replete and functional immune compartment. Within the immune compartment, T cells are often the effector arm of immune-based strategies due to their potent cytotoxic capabilities. However, many tumors have evolved a variety of mechanisms to evade T cell-mediated killing. Thus, while many T cell-based immunotherapies, such as immune checkpoint inhibition (ICI) and chimeric antigen receptor (CAR) T cells, have achieved considerable success in some solid cancers and hematological malignancies, these therapies often fail in solid tumors due to tumor-imposed T cell dysfunctions. These dysfunctional mechanisms broadly include reduced T cell access into and identification of tumors, as well as an overall immunosuppressive tumor microenvironment that elicits T cell exhaustion. Therefore, novel, rational approaches are necessary to overcome the barriers to T cell function elicited by solid tumors. In this review, we will provide an overview of conventional immunotherapeutic strategies and the various barriers to T cell anti-tumor function encountered in solid tumors that lead to resistance. We will also explore a sampling of emerging strategies specifically aimed to bypass these tumor-imposed boundaries to T cell-based immunotherapies., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Waibl Polania, Lerner, Wilkinson, Hoyt-Miggelbrink and Fecci.)
- Published
- 2021
- Full Text
- View/download PDF
8. Chemical library screens targeting an HIV-1 accessory factor/host cell kinase complex identify novel antiretroviral compounds.
- Author
-
Emert-Sedlak L, Kodama T, Lerner EC, Dai W, Foster C, Day BW, Lazo JS, and Smithgall TE
- Subjects
- Cell Line, Drug Evaluation, Preclinical, Enzyme Activation, HIV-1 physiology, High-Throughput Screening Assays, Humans, Molecular Structure, Protein Binding, Structure-Activity Relationship, Virus Replication, Anti-HIV Agents chemistry, Anti-HIV Agents pharmacology, HIV-1 drug effects, Proto-Oncogene Proteins c-hck metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, nef Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
Nef is an HIV-1 accessory protein essential for AIDS progression and an attractive target for drug discovery. Lack of a catalytic function makes Nef difficult to assay in chemical library screens. We developed a high-throughput screening assay for inhibitors of Nef function by coupling it to one of its host cell binding partners, the Src-family kinase Hck. Hck activation is dependent upon Nef in this assay, providing a direct readout of Nef activity in vitro. Using this screen, a unique diphenylfuropyrimidine was identified as a strong inhibitor of Nef-dependent Hck activation. This compound also exhibited remarkable antiretroviral effects, blocking Nef-dependent HIV replication in cell culture. Structurally related analogs were synthesized and shown to exhibit similar Nef-dependent antiviral activity, identifying the diphenylfuropyrimidine substructure as a new lead for antiretroviral drug development. This study demonstrates that coupling noncatalytic HIV accessory factors with host cell target proteins addressable by high-throughput assays may afford new avenues for the discovery of anti-HIV agents.
- Published
- 2009
- Full Text
- View/download PDF
9. HGF and c-Met participate in paracrine tumorigenic pathways in head and neck squamous cell cancer.
- Author
-
Knowles LM, Stabile LP, Egloff AM, Rothstein ME, Thomas SM, Gubish CT, Lerner EC, Seethala RR, Suzuki S, Quesnelle KM, Morgan S, Ferris RL, Grandis JR, and Siegfried JM
- Subjects
- Animals, Blotting, Western, Carcinoma, Squamous Cell pathology, Carcinoma, Squamous Cell physiopathology, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Crizotinib, Dose-Response Relationship, Drug, Head and Neck Neoplasms pathology, Head and Neck Neoplasms physiopathology, Hepatocyte Growth Factor pharmacology, Humans, Immunohistochemistry, Indoles pharmacology, Mice, Mice, Nude, Neoplasm Transplantation, Paracrine Communication physiology, Piperazines pharmacology, Piperidines pharmacology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Pyrazoles, Pyridines pharmacology, Signal Transduction drug effects, Stress, Mechanical, Sulfonamides pharmacology, Transplantation, Heterologous, Tumor Burden drug effects, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms metabolism, Hepatocyte Growth Factor metabolism, Proto-Oncogene Proteins c-met metabolism, Signal Transduction physiology
- Abstract
Purpose: We determined hepatocyte growth factor (HGF) and c-Met expression and signaling in human head and neck squamous cell carcinoma (HNSCC) cells and primary tissues and tested the ability of c-Met tyrosine kinase inhibitors (TKI) to block HGF-induced biological signaling., Experimental Design: Expression and signaling were determined using immunoblotting, ELISA, and immunohistochemistry. Biological end points included wound healing, cell proliferation, and invasion. c-Met TKIs were tested for their ability to block HGF-induced signaling and biological effects in vitro and in xenografts established in nude mice., Results: c-Met was expressed and functional in HNSCC cells. HGF was secreted by HNSCC tumor-derived fibroblasts, but not by HNSCC cells. Activation of c-Met promoted phosphorylation of AKT and mitogen-activated protein kinase as well as release of the inflammatory cytokine interleukin-8. Cell growth and wound healing were also stimulated by HGF. c-Met TKIs blocked HGF-induced signaling, interleukin-8 release, and wound healing. Enhanced invasion of HNSCC cells induced by the presence of tumor-derived fibroblasts was completely blocked with a HGF-neutralizing antibody. PF-2341066, a c-Met TKI, caused a 50% inhibition of HNSCC tumor growth in vivo with decreased proliferation and increased apoptosis within the tumors. In HNSCC tumor tissues, both HGF and c-Met protein were increased compared with expression in normal mucosa., Conclusions: These results show that HGF acts mainly as a paracrine factor in HNSCC cells, the HGF/c-Met pathway is frequently up-regulated and functional in HNSCC, and a clinically relevant c-Met TKI shows antitumor activity in vivo. Blocking the HGF/c-Met pathway may be clinically useful for the treatment of HNSCC.
- Published
- 2009
- Full Text
- View/download PDF
10. Functional characterization and conformational analysis of the Herpesvirus saimiri Tip-C484 protein.
- Author
-
Mitchell JL, Trible RP, Emert-Sedlak LA, Weis DD, Lerner EC, Applen JJ, Sefton BM, Smithgall TE, and Engen JR
- Subjects
- Mass Spectrometry, Peptides chemistry, Phosphoproteins genetics, Phosphoproteins isolation & purification, Phosphoproteins metabolism, Phosphorylation, Protein Conformation, Protons, Recombinant Proteins genetics, Viral Proteins genetics, Viral Proteins isolation & purification, Viral Proteins metabolism, Herpesvirus 2, Saimiriine enzymology, Phosphoproteins chemistry, Viral Proteins chemistry
- Abstract
Tyrosine kinase interacting protein (Tip) of Herpesvirus saimiri (HVS) activates the lymphoid-specific member of the Src family kinase Lck. The Tip:Lck interaction is essential for transformation and oncogenesis in HVS-infected cells. As there are no structural data for Tip, hydrogen-exchange mass spectrometry was used to investigate the conformation of a nearly full-length form (residues 1-187) of Tip from HVS strain C484. Disorder predictions suggested that Tip would be mostly unstructured, so great care was taken to ascertain whether recombinant Tip was functional. Circular dichroism and gel-filtration analysis indicated an extended, unstructured protein. In vitro and in vivo binding and kinase assays confirmed that purified, recombinant Tip interacted with Lck, was capable of activating Lck kinase activity strongly and was multiply phosphorylated by Lck. Hydrogen-exchange mass spectrometry of Tip then showed that the majority of backbone amide hydrogen atoms became deuterated after only 10 s of labeling. Such a result suggested that Tip was almost totally unstructured in solution. Digestion of deuterium-labeled Tip revealed some regions with minor protection from exchange. Overall, it was found that, although recombinant Tip is still functional and capable of binding and activating its target Lck, it is largely unstructured.
- Published
- 2007
- Full Text
- View/download PDF
11. C-terminal Src kinase-homologous kinase (CHK), a unique inhibitor inactivating multiple active conformations of Src family tyrosine kinases.
- Author
-
Chong YP, Chan AS, Chan KC, Williamson NA, Lerner EC, Smithgall TE, Bjorge JD, Fujita DJ, Purcell AW, Scholz G, Mulhern TD, and Cheng HC
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Catalysis, Cell Line, Down-Regulation, Enzyme Activation, Humans, Mutation genetics, Protein Binding, Protein Conformation, Spodoptera, src-Family Kinases classification, src-Family Kinases genetics, src-Family Kinases metabolism
- Abstract
The Src family of protein kinases (SFKs) mediates mitogenic signal transduction, and constitutive SFK activation is associated with tumorigenesis. To prevent constitutive SFK activation, the catalytic activity of SFKs in normal mammalian cells is suppressed mainly by two inhibitors called C-terminal Src kinase (CSK) and CSK-homologous kinase (CHK), which inactivate SFKs by phosphorylating a consensus tyrosine near the C terminus of SFKs (Y(T)). The phosphorylated Y(T) intramolecularly binds to the SH2 domain of SFKs. This interaction, known as pY(T)/SH2 interaction, together with binding between the SH2 kinase linker and the SH3 domain of SFKs (linker/SH3 interaction) stabilizes SFKs in a "closed" inactive conformation. We previously discovered an alternative mechanism CHK employs to inhibit SFKs. This mechanism, referred to as the non-catalytic inhibitory mechanism, involves tight binding of CHK to SFKs; the binding alone is sufficient to inhibit SFKs. Herein, we constructed multiple active conformations of an SFK member, Hck, by systematically disrupting the two inhibitory interactions. We found that CHK employs the non-catalytic mechanism to inactivate these active conformations of Hck. However, CHK does not bind Hck when it adopts the inactive conformation in which both inhibitory interactions are intact. These data indicate that binding of CHK to SFKs via the non-catalytic mechanism is governed by the conformations of SFKs. Although CSK is also an inhibitor of SFKs, it does not inhibit SFKs by a similar non-catalytic mechanism. Thus, the non-catalytic inhibitory mechanism is a unique property of CHK that allows it to down-regulate multiple active conformations of SFKs.
- Published
- 2006
- Full Text
- View/download PDF
12. Conformational features of the full-length HIV and SIV Nef proteins determined by mass spectrometry.
- Author
-
Hochrein JM, Wales TE, Lerner EC, Schiavone AP, Smithgall TE, and Engen JR
- Subjects
- Amino Acid Sequence, Deuterium Exchange Measurement, HIV chemistry, Mass Spectrometry, Molecular Sequence Data, Sequence Alignment, nef Gene Products, Human Immunodeficiency Virus, Gene Products, nef chemistry, Protein Conformation, Viral Regulatory and Accessory Proteins chemistry
- Abstract
The Nef protein from human or simian immunodeficiency virus enhances viral replication, downregulates immune cell receptors, and activates multiple host cell signaling pathways. Conformational information about full-length Nef has been difficult to obtain as the full-length protein is not readily amenable to NMR or X-ray crystallography due to aggregation at high concentrations. As an alternative, full-length HIV and SIV Nef were probed with hydrogen exchange mass spectrometry, a method compatible with the low concentration requirements of Nef. The results showed that HIV Nef contains a solvent-protected core, as previously demonstrated with both NMR and X-ray crystallography. SIV Nef, for which there is no structural information, had a similar protected core, although it was more flexible and dynamic than its HIV counterpart. Many of the regions outside the core in both SIV and HIV Nef were highly solvent exposed. However, limited protection from exchange was observed in both N- and C-terminal regions, suggesting the presence of structured elements. Protection from exchange was also observed in a large loop emanating from the core that was deleted for NMR and X-ray analysis. These data show that while the majority of Nef was highly solvent exposed, regions outside the core may have structural attributes which may contribute to Nef functions known to map to these regions.
- Published
- 2006
- Full Text
- View/download PDF
13. Phosphorylation of the varicella-zoster virus (VZV) major transcriptional regulatory protein IE62 by the VZV open reading frame 66 protein kinase.
- Author
-
Eisfeld AJ, Turse SE, Jackson SA, Lerner EC, and Kinchington PR
- Subjects
- Active Transport, Cell Nucleus, Amino Acid Substitution, Cell Nucleus metabolism, Immediate-Early Proteins chemistry, Mutation, Nuclear Localization Signals, Open Reading Frames, Phosphorus Radioisotopes metabolism, Phosphorylation, Sequence Deletion, Staining and Labeling, Substrate Specificity, Trans-Activators chemistry, Viral Envelope Proteins chemistry, Herpesvirus 3, Human metabolism, Immediate-Early Proteins metabolism, Protein Kinases metabolism, Trans-Activators metabolism, Viral Envelope Proteins metabolism, Viral Proteins metabolism
- Abstract
IE62, the major transcriptional regulatory protein encoded by varicella-zoster virus (VZV), is nuclear at early times of VZV infection but then becomes predominantly cytoplasmic as a result of expression of the protein kinase encoded by open reading frame 66 (ORF66). Cytoplasmic forms of IE62 are required for its inclusion as an abundant VZV virion tegument protein. Here we show that ORF66 directly phosphorylates IE62 at two residues, with phosphorylation at S686 being sufficient to regulate IE62 nuclear import. Phosphotryptic peptide analyses established an ORF66 kinase-mediated phosphorylation of the complete IE62 protein in transfected and VZV-infected cells. Using truncated and point-mutated IE62 peptides, ORF66-directed phosphorylation was mapped to residues S686 and S722, immediately downstream of the IE62 nuclear localization signal. An IE62 protein with an S686A mutation retained efficient nuclear import activity, even in the presence of functional ORF66 protein kinase, but an IE62 protein containing an S686D alteration was imported into the nucleus inefficiently. In contrast, the nuclear import of IE62 carrying an S722A mutation was still modulated by ORF66 expression, and IE62 with an S722D mutation was imported efficiently into the nucleus. An in vitro phosphorylation assay was developed using bacterially expressed IE62-maltose binding protein fusions as substrates for immunopurified ORF66 protein kinase from recombinant baculovirus-infected insect cells. ORF66 kinase phosphorylated the IE62 peptides, with similar specificities for residues S686 and S722. These results indicate that IE62 nuclear import is modulated as a result of direct phosphorylation of IE62 by ORF66 kinase. This represents an interaction that is, so far, unique among the alphaherpesviruses.
- Published
- 2006
- Full Text
- View/download PDF
14. An examination of dynamics crosstalk between SH2 and SH3 domains by hydrogen/deuterium exchange and mass spectrometry.
- Author
-
Hochrein JM, Lerner EC, Schiavone AP, Smithgall TE, and Engen JR
- Subjects
- Allosteric Site, Humans, Ligands, Mass Spectrometry, Peptide Fragments metabolism, Peptide Fragments physiology, Protein Binding, Proto-Oncogene Proteins c-hck chemistry, Proto-Oncogene Proteins c-hck metabolism, Proto-Oncogene Proteins c-hck physiology, Thermodynamics, Deuterium Exchange Measurement, Peptide Fragments chemistry, src Homology Domains physiology
- Abstract
The ability of proteins to regulate their own enzymatic activity can be facilitated by changes in structure or protein dynamics in response to external regulators. Because many proteins contain SH2 and SH3 domains, transmission of information between the domains is a potential method of allosteric regulation. To determine if ligand binding to one modular domain may alter structural dynamics in an adjacent domain, allowing potential transmission of information through the protein, we used hydrogen exchange and mass spectrometry to measure changes in protein dynamics in the SH3 and SH2 domains of hematopoietic cell kinase (Hck). Ligand binding to either domain had little or no effect on hydrogen exchange in the adjacent domain, suggesting that changes in protein structure or dynamics are not a means of SH2/SH3 crosstalk. Furthermore, ligands of varying affinity covalently attached to SH3/SH2 altered dynamics only in the domain to which they bind. Such results demonstrate that ligand binding may not structurally alter adjacent SH3/SH2 domains and implies that other aspects of protein architecture contribute to the multiple levels of regulation in proteins containing SH3 and SH2 domains.
- Published
- 2006
- Full Text
- View/download PDF
15. Activation of the Src family kinase Hck without SH3-linker release.
- Author
-
Lerner EC, Trible RP, Schiavone AP, Hochrein JM, Engen JR, and Smithgall TE
- Subjects
- Animals, Binding Sites, Biotinylation, Cell Line, Crystallography, X-Ray, Enzyme Activation, Gene Expression, Humans, Models, Molecular, Mutation, Peptide Fragments genetics, Protein Conformation, Protein Structure, Secondary, Proto-Oncogene Proteins c-hck chemistry, Proto-Oncogene Proteins c-hck genetics, Rats, Recombinant Proteins, Saccharomyces cerevisiae genetics, Sequence Analysis, DNA, Signal Transduction, Structure-Activity Relationship, Surface Plasmon Resonance, Transfection, Proto-Oncogene Proteins c-hck metabolism, src Homology Domains physiology
- Abstract
Src family protein-tyrosine kinases are regulated by intramolecular binding of the SH2 domain to the C-terminal tail and association of the SH3 domain with the SH2 kinase-linker. The presence of two regulatory interactions raises the question of whether disruption of both is required for kinase activation. To address this question, we engineered a high affinity linker (HAL) mutant of the Src family member Hck in which an optimal SH3 ligand was substituted for the natural linker. Surface plasmon resonance analysis demonstrated tight intramolecular binding of the modified HAL sequence to SH3. Hck-HAL was then combined with a tail tyrosine mutation (Y501F) and expressed in Rat-2 fibroblasts. Surprisingly, Hck-HAL-Y501F showed strong transforming and kinase activities, demonstrating that intramolecular SH3-linker release is not required for SH2-based kinase activation. In Saccharomyces cerevisiae, which lacks the negative regulatory tail kinase Csk, wild-type Hck was more strongly activated in the presence of an SH3-binding protein (human immunodeficiency virus-1 Nef), indicating persistence of native SH3-linker interaction in an active Hck conformation. Taken together, these data support the existence of multiple active conformations of Src family kinases that may generate unique downstream signals.
- Published
- 2005
- Full Text
- View/download PDF
16. The tyrosine kinases Fyn and Hck favor the recruitment of tyrosine-phosphorylated APOBEC3G into vif-defective HIV-1 particles.
- Author
-
Douaisi M, Dussart S, Courcoul M, Bessou G, Lerner EC, Decroly E, and Vigne R
- Subjects
- APOBEC-3G Deaminase, Binding Sites, Cytidine Deaminase, HeLa Cells, Humans, Kidney metabolism, Kidney virology, Monocytes metabolism, Monocytes virology, Nucleoside Deaminases, Phosphorylation, Protein Binding, Proto-Oncogene Proteins c-fyn, Proto-Oncogene Proteins c-hck, Repressor Proteins, T-Lymphocytes metabolism, T-Lymphocytes virology, Virion metabolism, vif Gene Products, Human Immunodeficiency Virus, Gene Products, vif metabolism, HIV-1 physiology, Protein-Tyrosine Kinases metabolism, Proteins metabolism, Proto-Oncogene Proteins metabolism, Tyrosine metabolism, src-Family Kinases metabolism
- Abstract
The main function of Vif is to limit the antiviral activity of APOBEC3G by counteracting its packaging into HIV-1 virions. In this work, we examine the possible functional interactions between Vif, APOBEC3G, and two Src family tyrosine kinases, Fyn and Hck, present in T lymphocytes and in monocyte-macrophages, respectively. By GST pull-down, we show that the SH3 domains of Fyn and Hck, and the corresponding full-length proteins bind Vif of HIV-1. One consequence of this interaction is a reduction in their catalytic activity. Interestingly, we also observed that APOBEC3G can be phosphorylated on tyrosine in the presence of Fyn or Hck, suggesting that both kinases may regulate APOBEC3G function. Accordingly, we demonstrate that in the presence of Fyn or Hck and in the absence of Vif, the overall level of APOBEC3G incorporated into HIV-1 particles is decreased, whereas the level of encapsidation of its phosphorylated form is significantly enhanced.
- Published
- 2005
- Full Text
- View/download PDF
17. Lyn regulates the cell death response to ultraviolet radiation through c-Jun N terminal kinase-dependent Fas ligand activation.
- Author
-
Shangary S, Lerner EC, Zhan Q, Corey SJ, Smithgall TE, and Baskaran R
- Subjects
- Animals, Antineoplastic Agents, Alkylating pharmacology, Cell Death physiology, Cell Death radiation effects, Cisplatin pharmacology, Down-Regulation physiology, Down-Regulation radiation effects, Eukaryotic Cells radiation effects, Fas Ligand Protein, Fetus, HeLa Cells, Humans, JNK Mitogen-Activated Protein Kinases, Membrane Glycoproteins radiation effects, Methyl Methanesulfonate pharmacology, Mice, Mice, Knockout, Mitogen-Activated Protein Kinases radiation effects, Radiation, Ionizing, src-Family Kinases radiation effects, Eukaryotic Cells enzymology, Membrane Glycoproteins metabolism, Mitogen-Activated Protein Kinases metabolism, Ultraviolet Rays adverse effects, src-Family Kinases metabolism
- Abstract
The Src-related tyrosine kinase, Lyn, plays an important role in mediating the cell cycle arrest and cell death response to genotoxic agents such as ionizing radiation. In this report we provide evidence to show that the catalytic function of Lyn is required for ultraviolet radiation (UV)- and methyl methanesulfonate (MMS)- but not for cisplatin (CDDP)- or ionizing radiation (IR)-induced cell death. Consequently, fibroblasts deficient in Lyn function were protected against cell death induction by UV and MMS, but showed normal cell death to IR and CDDP treatment. In Lyn(-/-) cells, UV-induced activation of stress-responsive kinases, Erk1/2 and p38, was normal; however, JNK activation was diminished. In addition, FasL induction by UV was also diminished in these cells. Reintroduction of wild-type Lyn restored JNK activation, FasL induction, and sensitivity to UV and MMS. A role for FasL in the cell death induction by Lyn-JNK signaling is indicated by the inhibition of cell death response by FasL neutralizing antibody. Together, the results support the presence of the Lyn-JNK signaling pathway that mediates the cell death response to UV and MMS treatment through FasL induction.
- Published
- 2003
- Full Text
- View/download PDF
18. Src kinases mediate STAT growth pathways in squamous cell carcinoma of the head and neck.
- Author
-
Xi S, Zhang Q, Dyer KF, Lerner EC, Smithgall TE, Gooding WE, Kamens J, and Grandis JR
- Subjects
- Base Sequence, Carcinoma, Squamous Cell enzymology, DNA Primers, Head and Neck Neoplasms enzymology, Humans, STAT3 Transcription Factor, STAT5 Transcription Factor, Tumor Cells, Cultured, src-Family Kinases physiology, Carcinoma, Squamous Cell pathology, Cell Division physiology, DNA-Binding Proteins physiology, Head and Neck Neoplasms pathology, Milk Proteins, Trans-Activators physiology, src-Family Kinases metabolism
- Abstract
Signal transducer and activator of transcription (STAT) proteins are constitutively activated in many malignancies, including squamous cell carcinoma of the head and neck (SCCHN). Previously, we reported that phosphorylation of the epidermal growth factor receptor (EGFR) is linked to activation of STATs 3 and 5 in SCCHN cells. The present study was undertaken to determine the role of Src family kinases in STAT activation and SCCHN growth. The Src family kinases c-Src, c-Yes, Fyn, and Lyn were expressed and activated by transforming growth factor-alpha stimulation in all four SCCHN cell lines examined but not in corresponding normal epithelial cells. In nine SCCHN cell lines tested, Src phosphotyrosine expression levels were highly correlated with activation levels of STATs 3 and 5. Co-immunoprecipitation analysis demonstrated interaction between c-Src and STATs 3 or 5 and EGFR in SCCHN cells, but no heterodimerization was detected between STAT3 and STAT5. SCCHN cells treated with either of two Src-specific inhibitors or transfected with a dominant-negative c-Src construct demonstrated decreased activation of STATs 3 and 5 and reduced growth rates in vitro. These results demonstrate a role for Src kinases in mediating activation of STATs 3 and 5 in concert with the EGFR in SCCHN cells. Strategies to target Src activation may contribute to the treatment of cancers that demonstrate increased levels of EGFR and STATs, including SCCHN.
- Published
- 2003
- Full Text
- View/download PDF
19. SH3-dependent stimulation of Src-family kinase autophosphorylation without tail release from the SH2 domain in vivo.
- Author
-
Lerner EC and Smithgall TE
- Subjects
- Animals, Cell Line, Enzyme Activation, Fibroblasts, Gene Products, nef metabolism, HIV-1, Humans, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Phosphorylation, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-hck, Signal Transduction, Spodoptera, Structure-Activity Relationship, nef Gene Products, Human Immunodeficiency Virus, src-Family Kinases genetics, Phosphotyrosine metabolism, Protein-Tyrosine Kinases chemistry, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins metabolism, src Homology Domains, src-Family Kinases chemistry, src-Family Kinases metabolism
- Abstract
Src family protein-tyrosine kinase activity is suppressed by two intramolecular interactions. These involve binding of the SH2 domain to the phosphorylated C-terminal tail and association of the SH3 domain with a polyproline type II helix formed by the SH2-kinase linker. Here we show that SH3-dependent activation of the Src family member Hck by HIV-1 Nef binding or by SH2-kinase linker mutation does not affect tail tyrosine phosphorylation in fibroblasts. Surprisingly, replacement of the wild type Hck tail with a high-affinity SH2 domain-binding sequence did not affect Hck activation or downstream signaling by these SH3-dependent mechanisms, suggesting that activation through SH3 occurs without SH2-tail dissociation. These results identify SH3-linker interaction as an independent mode of Hck kinase regulation in vivo and suggest that different mechanisms of Src kinase activation may generate distinct output signals because of differences in SH2 or SH3 domain accessibility.
- Published
- 2002
- Full Text
- View/download PDF
20. Differential function of STAT5 isoforms in head and neck cancer growth control.
- Author
-
Leong PL, Xi S, Drenning SD, Dyer KF, Wentzel AL, Lerner EC, Smithgall TE, and Grandis JR
- Subjects
- Carcinoma, Squamous Cell, Cell Division, Cell Line, Transformed, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, ErbB Receptors metabolism, Head and Neck Neoplasms, Humans, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Isoforms physiology, STAT5 Transcription Factor, Signal Transduction, Trans-Activators genetics, Trans-Activators metabolism, Transforming Growth Factor alpha metabolism, Tumor Cells, Cultured, Tumor Suppressor Proteins, DNA-Binding Proteins physiology, Milk Proteins, Trans-Activators physiology
- Abstract
Up-regulation of the epidermal growth factor receptor (EGFR) is critical for the loss of growth control in a variety of human cancers, including squamous cell carcinoma of the head and neck (SCCHN). Stimulation of EGFR results in activation of mitogenic signaling pathways including Signal Transducers and Activators of Transcription (STATs). Stat5 activation has been primarily demonstrated in hematopoietic malignancies. Gene disruption studies suggest potentially distinct functions of the Stat5 isoforms, Stat5a and Stat5b, which are encoded by two genes closely linked on human chromosome 17. To determine the function of Stat5 in SCCHN growth control, we studied the expression and constitutive activation of Stat5a and Stat5b in normal and transformed human squamous epithelial cells. Increased constitutive activation of Stat5 was detected in transformed compared with normal squamous cells. Blockade of TGF-alpha or EGFR, abrogated Stat5 activation. Targeting of Stat5b using antisense oligonucleotides inhibited SCCHN growth. In addition, SCCHN cells stably transfected with dominant negative mutant Stat5b failed to proliferate in vitro. In contrast, targeting of Stat5a using either antisense or dominant negative strategies had no effect on cell growth. These results suggest that TGF-alpha/EGFR-mediated autocrine growth of transformed epithelial cells is dependent on activation of Stat5b but not Stat5a.
- Published
- 2002
- Full Text
- View/download PDF
21. Control of myeloid differentiation and survival by Stats.
- Author
-
Smithgall TE, Briggs SD, Schreiner S, Lerner EC, Cheng H, and Wilson MB
- Subjects
- Animals, Antigens, CD genetics, Antigens, CD metabolism, Cell Survival, Cytokine Receptor gp130, DNA-Binding Proteins genetics, Hematopoiesis, Hematopoietic Stem Cells metabolism, Humans, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Receptors, Erythropoietin genetics, Receptors, Erythropoietin metabolism, Receptors, Granulocyte Colony-Stimulating Factor genetics, Receptors, Granulocyte Colony-Stimulating Factor metabolism, STAT3 Transcription Factor, STAT5 Transcription Factor, Signal Transduction, Trans-Activators genetics, src-Family Kinases metabolism, Cell Differentiation, DNA-Binding Proteins metabolism, Hematopoietic Stem Cells cytology, Milk Proteins, Trans-Activators metabolism
- Abstract
Hematopoiesis involves a complex array of growth factors that regulate the survival and proliferation of immature progenitors, influence differentiation commitment, and modulate end-stage cell functions. This mini-review is focused on the role of Stat activation in the development of myeloid cells in response to hematopoietic cytokines. Much of the evidence implicating Stats in these cellular processes comes from studies of mutant cytokine receptors selectively uncoupled from Stat activation, dominant-inhibitory Stat mutants, and mice with targeted disruptions of Stat genes. Together these approaches provide strong evidence that Stat activation, particularly of Stat3 and Stat5, plays an important role in myeloid differentiation and survival. Oncogene (2000).
- Published
- 2000
- Full Text
- View/download PDF
22. The nonreceptor protein-tyrosine kinase c-Fes is involved in fibroblast growth factor-2-induced chemotaxis of murine brain capillary endothelial cells.
- Author
-
Kanda S, Lerner EC, Tsuda S, Shono T, Kanetake H, and Smithgall TE
- Subjects
- Animals, Brain blood supply, Capillaries, Cell Division, Cell Line, Cerebrovascular Circulation drug effects, Chemotaxis drug effects, Endothelium, Vascular drug effects, Humans, Mice, Mice, Transgenic, Plasminogen Activators metabolism, Proto-Oncogene Proteins c-fes, Recombinant Proteins metabolism, Signal Transduction, Transfection, Cerebrovascular Circulation physiology, Chemotaxis physiology, Endothelium, Vascular physiology, Fibroblast Growth Factor 2 pharmacology, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Fibroblast growth factor-2 (FGF-2)-induced migration of endothelial cells is involved in angiogenesis in vivo. However, signal transduction pathways leading to FGF-2-induced chemotaxis of endothelial cells are largely unknown. Previous studies have shown that the cytoplasmic protein-tyrosine kinase c-Fes is expressed in vascular endothelial cells and may influence angiogenesis in vivo. To investigate the contribution of c-Fes to FGF-2 signaling, we expressed wild-type or kinase-inactive human c-Fes in the murine brain capillary endothelial cell line, IBE (Immortomouse brain endothelial cells). Wild-type c-Fes was tyrosine-phosphorylated upon FGF-2-stimulation in transfected cells, whereas kinase-inactive c-Fes was not. Overexpression of wild-type c-Fes promoted FGF-2-independent tube formation of IBE cells. Tube formation was not observed with endothelial cells expressing kinase-inactive c-Fes, indicating a requirement for c-Fes kinase activity in this biological response. Expression of kinase-defective c-Fes suppressed endothelial cell migration following FGF-2 treatment, suggesting that activation of endogenous c-Fes may be required for the chemotactic response. Expression of either wild-type c-Fes or the kinase-inactive mutant did not affect the tyrosine phosphorylation FRS2, Shc, or phospholipase C-gamma, nor did it influence the kinetics of mitogen-activated protein kinase activation. These results implicate c-Fes in FGF-2-induced chemotaxis of endothelial cells through signaling pathways not linked to mitogenesis.
- Published
- 2000
- Full Text
- View/download PDF
23. Affinity of Src family kinase SH3 domains for HIV Nef in vitro does not predict kinase activation by Nef in vivo.
- Author
-
Briggs SD, Lerner EC, and Smithgall TE
- Subjects
- Animals, Binding Sites, Cell Line, Cell Line, Transformed, HIV-1 physiology, Humans, Protein-Tyrosine Kinases chemistry, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins c-hck, Rats, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Spodoptera, Transfection, Virus Replication, nef Gene Products, Human Immunodeficiency Virus, src Homology Domains, Gene Products, nef metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, src-Family Kinases chemistry, src-Family Kinases metabolism
- Abstract
Nef is an HIV accessory protein required for high-titer viral replication and AIDS progression. Previous studies have shown that the SH3 domains of Hck and Lyn bind to Nef via proline-rich sequences in vitro, identifying these Src-related kinases as potential targets for Nef in vivo. Association of Nef with Hck causes displacement of the intramolecular interaction between the SH3 domain and the SH2-kinase linker, leading to kinase activation both in vitro and in vivo. In this study, we investigated whether interaction with Nef induces activation of other Src family kinases (Lyn, Fyn, Src, and Lck) following coexpression with Nef in Rat-2 fibroblasts. Coexpression with Nef induced Hck kinase activation and fibroblast transformation, consistent with previous results. In contrast, coexpression of Nef with Lyn was without effect, despite equivalent binding of Nef to full-length Lyn and Hck. Furthermore, Nef was found to suppress the kinase and transforming activities of Fyn, the SH3 domain of which exhibits low affinity for Nef. Coexpression with Nef did not alter c-Src or Lck tyrosine kinase or transforming activity in this system. Differential modulation of Src family members by Nef may produce unique downstream signals depending on the profile of Src kinases expressed in a given cell type.
- Published
- 2000
- Full Text
- View/download PDF
24. Inhibition of the prenylation of K-Ras, but not H- or N-Ras, is highly resistant to CAAX peptidomimetics and requires both a farnesyltransferase and a geranylgeranyltransferase I inhibitor in human tumor cell lines.
- Author
-
Lerner EC, Zhang TT, Knowles DB, Qian Y, Hamilton AD, and Sebti SM
- Subjects
- Benzamides pharmacology, Cell Division drug effects, Cell Division genetics, Farnesyltranstransferase, GTP-Binding Proteins drug effects, GTP-Binding Proteins immunology, GTP-Binding Proteins metabolism, Humans, Methionine analogs & derivatives, Methionine pharmacology, Peptides pharmacology, Protein Prenylation drug effects, Tumor Cells, Cultured, rap GTP-Binding Proteins, ras Proteins genetics, ras Proteins immunology, Alkyl and Aryl Transferases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Genes, ras, ras Proteins metabolism
- Abstract
The farnesyltransferase (FTase) inhibitor FTI-277 is highly effective at blocking oncogenic H-Ras but not K-Ras4B processing and signaling. While inhibition of processing and signaling of oncogenic K-Ras4B is more sensitive to the geranylgeranyltransferase I (GGTase I) inhibitor GGTI-286 than it is to FTI-277 in K-Ras4B-transformed NIH3T3 cells, the sensitivity of K-Ras as well as H- and N-Ras to the CAAX peptidomimetics in human tumor cell lines is not known. Here, we report that a panel of five human carcinoma cell lines from pancreatic, pulmonary, and bladder origins all express H-, N-, and K-Ras, and their respective prenylation sensitivities to the FTase and GGTase I inhibitors is variable. In all of the cell lines investigated, the prenylation of N-Ras was highly sensitive to FTI-277, and in two of the cell lines, N-Ras showed slight sensitivity to GGTI-298, an analog of GGTI-286. Although the prenylation of H-Ras was also sensitive to FTI-277, complete inhibition of H-Ras processing even at high concentrations of FTI-277 and/or GGTI-298 was never achieved. The prenylation of K-Ras, on the other hand, was highly resistant to FTI-277 and GGTI-298. Most significantly, treatment of human tumor cell lines with both inhibitors was required for inhibition of K-Ras prenylation. In one cell line, the human lung adenocarcinoma A-549, prenylation of K-Ras was highly resistant even when co-treated with both inhibitors. Furthermore, soft agar experiments demonstrated that in all the human tumor cell lines tested inhibition of K-Ras prenylation was not necessary for inhibition of anchorage-independent growth. In addition, although GGTI-298 had very little effect on soft agar growth, the combination of FTI-277 and GGTI-298 resulted in significant growth inhibition. Therefore, the results demonstrate that while FTI-277 inhibits N-Ras and H-Ras processing in the human tumor cell lines evaluated, inhibition of K-Ras processing requires both an FTase inhibitor as well as a GGTase I inhibitor, and that inhibition of human tumor growth in soft agar does not require inhibition of oncogenic K-Ras processing.
- Published
- 1997
- Full Text
- View/download PDF
25. Inhibition of Ras prenylation: a signaling target for novel anti-cancer drug design.
- Author
-
Lerner EC, Hamilton AD, and Sebti SM
- Subjects
- Animals, Farnesyltranstransferase, Humans, Mice, Transferases antagonists & inhibitors, Alkyl and Aryl Transferases, Antineoplastic Agents pharmacology, Dimethylallyltranstransferase antagonists & inhibitors, Drug Design, Protein Prenylation drug effects, Signal Transduction drug effects, ras Proteins drug effects
- Abstract
The cancer-causing activity of Ras requires the prenylation of a cysteine fourth from its carboxyl terminus. Rational design of peptidomimetics of the carboxyl terminal tetrapeptide prenylation site on Ras resulted in pharmacological agents capable of inhibiting Ras processing, selectively antagonizing oncogenic signaling and suppressing human tumor growth in mouse models without side effects. This mini-review describes the efforts of several groups to design, synthesize and evaluate the biological activities of farnesyltransferase and geranylgeranyltransferase I inhibitors. Among the important issues that will be discussed are the mechanism of action of these inhibitors and the potential mechanisms of resistance to inhibition of K-Ras farnesylation.
- Published
- 1997
26. Ras CAAX peptidomimetic FTI-277 selectively blocks oncogenic Ras signaling by inducing cytoplasmic accumulation of inactive Ras-Raf complexes.
- Author
-
Lerner EC, Qian Y, Blaskovich MA, Fossum RD, Vogt A, Sun J, Cox AD, Der CJ, Hamilton AD, and Sebti SM
- Subjects
- Amino Acid Sequence, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Line, Cell Transformation, Neoplastic drug effects, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Farnesyltranstransferase, Humans, Methionine chemical synthesis, Methionine chemistry, Methionine pharmacology, Molecular Sequence Data, Oligopeptides chemical synthesis, Oligopeptides chemistry, Protein Processing, Post-Translational drug effects, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-raf, Signal Transduction drug effects, Transferases antagonists & inhibitors, ras Proteins genetics, Alkyl and Aryl Transferases, Enzyme Inhibitors pharmacology, Methionine analogs & derivatives, Oligopeptides pharmacology, ras Proteins metabolism
- Abstract
Ras-induced malignant transformation requires Ras farnesylation, a lipid posttranslational modification catalyzed by farnesyltransferase (FTase). Inhibitors of this enzyme have been shown to block Ras-dependent transformation, but the mechanism by which this occurs remains largely unknown. We have designed FTI-276, a peptide mimetic of the COOH-terminal Cys-Val-Ile-Met of K-Ras4B that inhibited potently FTase in vitro (IC50 = 500 pM) and was highly selective for FTase over geranylgeranyltransferase I (GGTase I) (IC50 = 50 nM). FTI-277, the methyl ester derivative of FTI-276, was extremely potent (IC50 = 100 nM) at inhibiting H-Ras, but not the geranylgeranylated Rap1A processing in whole cells. Treatment of H-Ras oncogene-transformed NIH 3T3 cells with FTI-277 blocked recruitment to the plasma membrane and subsequent activation of the serine/threonine kinase c-Raf-1 in cells transformed by farnesylated Ras (H-RasF), but not geranylgeranylated, Ras (H-RasGG). FTI-277 induced accumulation of cytoplasmic non-farnesylated H-Ras that was able to bind Raf and form cytoplasmic Ras/Raf complexes in which Raf kinase was not activated. Furthermore, FTI-277 blocked constitutive activation of mitogen-activated protein kinase (MAPK) in H-RasF, but not H-RasGG, or Raf-transformed cells. FTI-277 also inhibited oncogenic K-Ras4B processing and constitutive activation of MAPK, but the concentrations required were 100-fold higher than those needed for H-Ras inhibition. The results demonstrate that FTI-277 blocks Ras oncogenic signaling by accumulating inactive Ras/Raf complexes in the cytoplasm, hence preventing constitutive activation of the MAPK cascade.
- Published
- 1995
- Full Text
- View/download PDF
27. Disruption of oncogenic K-Ras4B processing and signaling by a potent geranylgeranyltransferase I inhibitor.
- Author
-
Lerner EC, Qian Y, Hamilton AD, and Sebti SM
- Subjects
- 3T3 Cells, Animals, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Humans, Leucine pharmacology, Mice, Protein Prenylation drug effects, Protein Processing, Post-Translational drug effects, Signal Transduction drug effects, Alkyl and Aryl Transferases, Enzyme Inhibitors pharmacology, Leucine analogs & derivatives, Transferases antagonists & inhibitors, ras Proteins metabolism
- Abstract
Prenylation of the carboxyl-terminal CAAX (C, cysteine; A, aliphatic acid; and X, any amino acid) of Ras is required for its biological activity. We have designed a CAAX peptidomimetic, GGTI-287, which is 10 times more potent toward inhibiting geranylgeranyltransferase I (GGTase I) in vitro (IC50 = 5 nM) than our previously reported farnesyltransferase inhibitor, FTI-276. In whole cells, the methyl ester derivative of GGTI-287, GGTI-286, was 25-fold more potent (IC50 = 2 microM) than the corresponding methyl ester of FTI-276, FTI-277, toward inhibiting the processing of the geranylgeranylated protein Rap1A. Furthermore, GGTI-286 is highly selective for geranylgeranylation over farnesylation since it inhibited the processing of farnesylated H-Ras only at much higher concentrations (IC50 > 30 microM). While the processing of H-Ras was very sensitive to inhibition by FTI-277 (IC50 = 100 nM), that of K-Ras4B was highly resistant (IC50 = 10 microM). In contrast, we found the processing of K-Ras4B to be much more sensitive to GGTI-286 (IC50 = 2 microM). Furthermore, oncogenic K-Ras4B stimulation inhibited potently by GGTI-286 (IC50 = 1 microM) but weakly by FTI-277 (IC50 = 30 microM). Significant inhibition of oncogenic K-Ras4B stimulation of MAP kinase by GGTI-286 occurred at concentrations (1-3 microM) that did not inhibit oncogenic H-Ras stimulation of MAP kinase. The data presented in this study provide the first demonstration of selective disruption of oncogenic K-Ras4B processing and signaling by a CAAX peptidomimetic. The higher sensitivity of K-Ras4B toward a GGTase I inhibitor has a tremendous impact on future research directions targeting Ras in anticancer therapy.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.