Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Matrix Biol ; 108: 20-38, 2022 04.
Article in English | MEDLINE | ID: mdl-35227929

ABSTRACT

The extracellular matrix molecule Tenascin-C (TNC) promotes cancer and chronic inflammation by multiple mechanisms. Recently, TNC was shown to promote an immune suppressive tumor microenvironment (TME) through binding soluble chemoattracting factors, thus retaining leukocytes in the stroma. TNC also binds to fibronectin (FN) and other molecules, raising the question of a potential common TNC binding mechanism. By sequence comparison of two TNC-interacting domains in FN, the fifth (FN5) and thirteenth (FN13) fibronectin type III domains we identified a MAtrix REgulating MOtif "MAREMO" or M-motif that is highly conserved amongst vertebrates. By sequence analysis, structural modeling and functional analysis we found also putative M-motifs in TNC itself. We showed by negative staining electron microscopic imaging that the M-motif in FN mediates interactions with FN as well as with TNC. We generated two M-motif mimetic peptides P5 and P13 resembling the M-motif in FN5 and FN13, respectively. By using structural information we modelled binding of these M-motif mimetics revealing a putative MAREMO binding site MBS in FN5 and TN3, respectively overlapping with the M-motif. We further demonstrated that the M-motif mimetic peptides blocked several functions of TNC, such as binding of TNC to FN, cell rounding on a mixed FN/TNC substratum, FN matrix expression and subsequent assembly, TNC-induced signaling and gene expression, TNC chemokine binding and dendritic cell retention, thus providing novel opportunities to inhibit TNC actions. Our results suggest that targeting the MAREMO/MBS interaction could be exploited for reducing inflammation and matrix functions in cancer and fibrosis.


Subject(s)
Neoplasms , Tenascin , Animals , Extracellular Matrix/metabolism , Inflammation , Neoplasms/genetics , Peptides , Tenascin/genetics , Tenascin/metabolism , Tumor Microenvironment
2.
EMBO Mol Med ; 13(6): e13270, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33988305

ABSTRACT

Immune checkpoint therapy, where CD8 tumor infiltrating T lymphocytes (TIL) are reactivated, is a promising anti-cancer treatment approach, yet with low response rates. The extracellular matrix, in particular tenascin-C, may generate barriers for TIL. To investigate this possibility, we used a MMTV-NeuNT and syngeneic mammary gland grafting model derived thereof with engineered tenascin-C levels and observed accumulation of CD8 TIL in tenascin-C-rich stroma. Inhibition studies revealed that tenascin-C induced CXCL12 through TLR4. By binding CXCL12, tenascin-C retained CD8 TIL in the stroma. Blockade of CXCR4, the receptor of CXCL12, enhanced macrophage and CD8 TIL infiltration and reduced tumor growth and subsequent metastasis. Retention of CD8 TIL by tenascin-C/CXCL12 was also observed in human breast cancer by tissue staining. Moreover, whereas high CD8 TIL numbers correlated with longer metastasis-free survival, this was not the case when also tenascin-C and CXCL12 levels were high. Altogether, these results may be useful for improving tumor immunity as diagnostic tool and to formulate a future "TIL-matrix-release-and-reactivate" strategy.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Neoplasms , CD8-Positive T-Lymphocytes , Chemokine CXCL12 , Extracellular Matrix , Humans , Tenascin
3.
Cancer Immunol Res ; 8(9): 1122-1138, 2020 09.
Article in English | MEDLINE | ID: mdl-32665262

ABSTRACT

Inherent immune suppression represents a major challenge in the treatment of human cancer. The extracellular matrix molecule tenascin-C promotes cancer by multiple mechanisms, yet the roles of tenascin-C in tumor immunity are incompletely understood. Using a 4NQO-induced oral squamous cell carcinoma (OSCC) model with abundant and absent tenascin-C, we demonstrated that tenascin-C enforced an immune-suppressive lymphoid stroma via CCL21/CCR7 signaling, leading to increased metastatic tumors. Through TLR4, tenascin-C increased expression of CCR7 in CD11c+ myeloid cells. By inducing CCL21 in lymphatic endothelial cells via integrin α9ß1 and binding to CCL21, tenascin-C immobilized CD11c+ cells in the stroma. Inversion of the lymph node-to-tumor CCL21 gradient, recruitment of T regulatory cells, high expression of anti-inflammatory cytokines, and matrisomal components were hallmarks of the tenascin-C-instructed lymphoid stroma. Ablation of tenascin-C or CCR7 blockade inhibited the lymphoid immune-suppressive stromal properties, reducing tumor growth, progression, and metastasis. Thus, targeting CCR7 could be relevant in human head and neck tumors, as high tenascin-C expression and an immune-suppressive stroma correlate to poor patient survival.


Subject(s)
Mouth Neoplasms/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Tenascin/immunology , Animals , Chemokine CCL21/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouth Neoplasms/pathology , Receptors, CCR7/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Tenascin/pharmacology , Tumor Microenvironment/immunology
4.
Crit Care Explor ; 1(9): e0044, 2019 Sep.
Article in English | MEDLINE | ID: mdl-32166286

ABSTRACT

Care-related infections affect up to 11% of ICU patients. Running therapeutic albumin is sometimes associated to less infection: whether a specific method of its infusion is of any interest to modulate innate defense is unknown. Our objectives were: 1) to test whether the method for albumin infusion is important to prevent care-related infections and 2) to analyze in vitro the antioxidative role of albumin on host defense proteins during shock (using vasostatin-I as an example). DESIGN: In a prospective, randomized, open-label trial, shock patients were allocated to receive either continuously 4% albumin or intermittently 20% albumin, as long as they were infused with norepinephrine. A translational study including in vivo and in vitro analyses of albumin-vasostatin-I interactions is reported. SETTING: A tertiary ICU caring for 1,000 patients per year. PATIENTS: Fifty shock patients with serum albumin less than 20 g/L. INTERVENTIONS: In vivo colonization and nosocomial infections were recorded and time-dependent changes in serum albumin, chromogranin A, and vasostatin-I concentrations as well. In vitro, we studied biochemical albumin-vasostatin-I relationship using biochemical methods. MEASUREMENTS AND MAIN RESULTS: Over 18 days, we recorded a decrease in colonization (four vs 12 episodes; p = 0.035) and nosocomial infection frequency (two vs 13 episodes; p = 0.002) in patients infused continuously 4% albumin versus controls. In vitro, albumin interacts with the disulfide loop vasostatin-I (residues 17-40) and continuous 4% albumin infusion restores its oxidative status required for antimicrobial activity. CONCLUSIONS: Continuous 4% albumin is effective in reducing care-related infections in shock patients by increasing the availability of antimicrobial vasostatin-I. This might guide future care of shock patients.

5.
Biol Cell ; 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29907957

ABSTRACT

BACKGROUND INFORMATION: Tumor stroma remodeling is a key feature of malignant tumors and can promote cancer progression. Laminins are major constituents of basement membranes that physically separate the epithelium from the underlying stroma. RESULTS: By employing mouse models expressing high and low levels of the laminin α1 chain (LMα1), we highlighted its implication in a tumor-stroma crosstalk, thus leading to increased colon tumor incidence, angiogenesis and tumor growth. The underlying mechanism involves attraction of carcinoma-associated fibroblasts by LMα1, VEGFA expression triggered by the complex integrin α2ß1-CXCR4 and binding of VEGFA to LM-111, which in turn promotes angiogenesis, tumor cell survival and proliferation. A gene signature comprising LAMA1, ITGB1, ITGA2, CXCR4 and VEGFA has negative predictive value in colon cancer. CONCLUSIONS: Together, we have identified VEGFA, CXCR4 and α2ß1 integrin downstream of LMα1 in colon cancer as of bad prognostic value for patient survival. SIGNIFICANCE: This information opens novel opportunities for diagnosis and treatment of colon cancer.

6.
Oncotarget ; 7(36): 57851-57865, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27506939

ABSTRACT

The neuropilin-plexin receptor complex regulates tumor cell migration and proliferation and thus is an interesting therapeutic target. High expression of neuropilin-1 is indeed associated with a bad prognosis in glioma patients. Q-RTPCR and tissue-array analyses showed here that Plexin-A1 is highly expressed in glioblastoma and that the highest level of expression correlates with the worse survival of patients. We next identified a developmental and tumor-associated pro-angiogenic role of Plexin-A1. Hence, by using molecular simulations and a two-hybrid like assay in parallel with biochemical and cellular assays we developed a specific Plexin-A1 peptidic antagonist disrupting transmembrane domain-mediated oligomerization of the receptor and subsequent signaling and functional activity. We found that this peptide exhibits anti-tumor activity in vivo on different human glioblastoma models including glioma cancer stem cells. Thus, screening Plexin-A1 expression and targeting Plexin-A1 in glioblastoma patients exhibit diagnostic and therapeutic value.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioma/pathology , Neovascularization, Pathologic/prevention & control , Nerve Tissue Proteins/antagonists & inhibitors , Peptides/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Animals , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chick Embryo , Chorioallantoic Membrane/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Nerve Tissue Proteins/metabolism , Protein Domains , Receptors, Cell Surface/metabolism , Tissue Array Analysis , Zebrafish
7.
Oncotarget ; 7(34): 54723-54732, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27351129

ABSTRACT

The transmembrane domains (TMD) in membrane receptors play a key role in cell signaling. As previously shown by us a peptide targeting the TMD of neuropilin-1 (MTP-NRP1), blocks cell proliferation, cell migration and angiogenesis in vitro, and decreases glioblastoma growth in vivo. We now explored the clinical potential of MTP-NRP1 on breast cancer models and demonstrate that MTP-NRP1 blocks proliferation of several breast cancer lines including the MDA-MB-231, a triple negative human breast cancer cell line. In models with long term in vivo administration of the peptide, MTP-NRP1 not only reduced tumor volume but also decreased number and size of breast cancer metastases. Strikingly, treating mice before tumors developed protected from metastasis establishment/formation. Overall, our results report that targeting the TMD of NRP1 in breast cancer is a potent new strategy to fight against breast cancer and related metastasis.


Subject(s)
Breast Neoplasms/drug therapy , Neuropilin-1/chemistry , Peptides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Neoplasm Metastasis
8.
Front Pharmacol ; 6: 283, 2015.
Article in English | MEDLINE | ID: mdl-26635612

ABSTRACT

Growth factors mediate their diverse biologic responses (regulation of cellular proliferation, differentiation, migration and survival) by binding to and activating cell-surface receptors with intrinsic protein kinase activity named receptor tyrosine kinases (RTKs). About 60 RTKs have been identified and can be classified into more than 16 different receptor families. Their activity is normally tightly controlled and regulated. Overexpression of RTK proteins or functional alterations caused by mutations in the corresponding genes or abnormal stimulation by autocrine growth factor loops contribute to constitutive RTK signaling, resulting in alterations in the physiological activities of cells. The ErbB receptor family of RTKs comprises four distinct receptors: the EGFR (also known as ErbB1/HER1), ErbB2 (neu, HER2), ErbB3 (HER3) and ErbB4 (HER4). ErbB family members are often overexpressed, amplified, or mutated in many forms of cancer, making them important therapeutic targets. EGFR has been found to be amplified in gliomas and non-small-cell lung carcinoma while ErbB2 amplifications are seen in breast, ovarian, bladder, non-small-cell lung carcinoma, as well as several other tumor types. Several data have shown that ErbB receptor family and its downstream pathway regulate epithelial-mesenchymal transition, migration, and tumor invasion by modulating extracellular matrix (ECM) components. Recent findings indicate that ECM components such as matrikines bind specifically to EGF receptor and promote cell invasion. In this review, we will present an in-depth overview of the structure, mechanisms, cell signaling, and functions of ErbB family receptors in cell adhesion and migration. Furthermore, we will describe in a last part the new strategies developed in anti-cancer therapy to inhibit ErbB family receptor activation.

9.
J Mol Biol ; 426(24): 4099-4111, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25315821

ABSTRACT

Signaling in eukaryotic cells frequently relies on dynamic interactions of single-pass membrane receptors involving their transmembrane (TM) domains. To search for new such interactions, we have developed a bacterial two-hybrid system to screen for both homotypic and heterotypic interactions between TM helices. We have explored the dimerization of TM domains from 16 proteins involved in both receptor tyrosine kinase and neuropilin signaling. This study has revealed several new interactions. We found that the TM domain of Mucin-4, a putative intramembrane ligand for erbB2, dimerizes not only with erbB2 but also with all four members of the erbB family. In the Neuropilin/Plexin family of receptors, we showed that the TM domains of Neuropilins 1 and 2 dimerize with themselves and also with Plexin-A1, Plexin-B1, and L1CAM, but we were unable to observe interactions with several other TM domains notably those of members of the VEGF receptor family. The potentially important Neuropilin 1/Plexin-A1 interaction was confirmed using a surface plasmon resonance assay. This work shows that TM domain interactions can be highly specific. Exploring further the propensities of TM helix-helix association in cell membrane should have important practical implications related to our understanding of the structure-function of bitopic proteins' assembly and subsequent function, especially in the regulation of signal transduction.


Subject(s)
Cell Membrane/metabolism , Neuropilin-1/metabolism , Neuropilin-2/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Blotting, Western , Humans , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Neuropilin-1/chemistry , Neuropilin-1/genetics , Neuropilin-2/chemistry , Neuropilin-2/genetics , Protein Binding , Protein Interaction Mapping/methods , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
10.
Cell Rep ; 8(6): 1714-1721, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25220456

ABSTRACT

Breast cancer is still a deadly disease despite major achievements in targeted therapies designed to block ligands or ligand-binding subunits of major tyrosine kinase receptors. Relapse is significant and metastases deleterious, which demands novel strategies for fighting this disease. Here, we report a proof-of-concept experiment demonstrating that small peptides interfering with the transmembrane domain of the tyrosine kinase epidermal growth factor receptor ErbB2 exhibit anticancer properties when used at micromolar dosages in a genetically engineered mouse model of breast cancer. Different assays demonstrate the specificity of the ErbB2-targeting peptide, which induces long-term reduction of ErbB2 phosphorylation and Akt signaling consistent with reduced tumor cell proliferation and increased survival. Microcomputed tomography analysis established the antimetastatic activity of the peptide and its impact on primary tumor growth. This reveals the interior of the cell membrane as an unexplored dimension for drug design.


Subject(s)
Receptor, ErbB-2/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Transgenic , Molecular Sequence Data , Peptides/chemistry , Peptides/therapeutic use , Peptides/toxicity , Phosphorylation/drug effects , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Tomography, X-Ray Computed
11.
Cell Rep ; 5(2): 482-92, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139798

ABSTRACT

The extracellular matrix molecule tenascin-C (TNC) is a major component of the cancer-specific matrix, and high TNC expression is linked to poor prognosis in several cancers. To provide a comprehensive understanding of TNC's functions in cancer, we established an immune-competent transgenic mouse model of pancreatic ß-cell carcinogenesis with varying levels of TNC expression and compared stochastic neuroendocrine tumor formation in abundance or absence of TNC. We show that TNC promotes tumor cell survival, the angiogenic switch, more and leaky vessels, carcinoma progression, and lung micrometastasis. TNC downregulates Dickkopf-1 (DKK1) promoter activity through the blocking of actin stress fiber formation, activates Wnt signaling, and induces Wnt target genes in tumor and endothelial cells. Our results implicate DKK1 downregulation as an important mechanism underlying TNC-enhanced tumor progression through the provision of a proangiogenic tumor microenvironment.


Subject(s)
Down-Regulation , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Tenascin/metabolism , Wnt Proteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Signal Transduction , Tenascin/deficiency , Tenascin/genetics , Wnt Proteins/antagonists & inhibitors
12.
Cell Adh Migr ; 3(4): 383-9, 2009.
Article in English | MEDLINE | ID: mdl-19855168

ABSTRACT

Semaphorin 3A (Sema3A) is a secreted guidance molecule initially described in the nervous system. This protein is able to control axon growth but also effects on endothelial cells migration. Here, we report that Sema3A acts as a chemorepellent factor for the rat C6 glioma cells and three different human glioma cell lines. Interestingly, Sema3A triggered a chemoattractive response in a fourth human glioma cell line. The nature of the receptor complex ensuring the appropriate signaling was dissected in C6 cells by using function blocking antibodies and gain- or loss-of function experiments using recombinant receptors. Our results demonstrate that neuropilin-1, neuropilin-2 and PlexinA1 are necessary to trigger cell repulsion. The selective blockade of neuropilin-1 or Plexin-A1 switched the chemorepulsive effect of Sema3A into a chemoattractive one. Strikingly, blocking Neuropilin-2 suppressed Sema3A-induced cell migration while overexpression of neuropilin-2 was able to convert the chemorepulsive effect of Sema3A into a chemoattractive one. Our results not only provide additional evidence for a biological function of Sema3A in glioma migration but also reveal part of the receptor complex involved. Hence, our study describes a receptor-based plasticity in cancer cells leading to opposite migration behavior in response to the same extracellular signal.


Subject(s)
Cell Movement/physiology , Glioma/pathology , Neuropilin-2/physiology , Semaphorin-3A/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/genetics , Neuropilin-1/immunology , Neuropilin-1/metabolism , Neuropilin-2/antagonists & inhibitors , Rats , Receptors, Cell Surface/metabolism , Semaphorin-3A/genetics , Transfection
13.
Mol Biol Cell ; 19(2): 646-54, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18045991

ABSTRACT

Neuropilin-1 (NRP1) is a transmembrane receptor playing a pivotal role in the control of semaphorins and VEGF signaling pathways. The exact mechanism controlling semaphorin receptor complex formation is unknown. A structural analysis and modeling of NRP1 revealed a putative dimerization GxxxG motif potentially important for NRP1 dimerization and oligomerization. Our data show that this motif mediates the dimerization of the transmembrane domain of NRP1 as demonstrated by a dimerization assay (ToxLuc assay) performed in natural membrane and FRET analysis. A synthetic peptide derived from the transmembrane segment of NRP1 abolished the inhibitory effect of Sema3A. This effect depends on the capacity of the peptide to interfere with NRP1 dimerization and the formation of oligomeric complexes. Mutation of the GxxxG dimerization motif in the transmembrane domain of NRP1 confirmed its biological importance for Sema3A signaling. Overall, our results shed light on an essential step required for semaphorin signaling and provide novel evidence for the crucial role of transmembrane domain of bitopic protein containing GxxxG motif in the formation of receptor complexes that are a prerequisite for cell signaling.


Subject(s)
Neuropilin-1/chemistry , Neuropilin-1/metabolism , Amino Acid Motifs , Animals , COS Cells , Centrifugation, Density Gradient , Chlorocebus aethiops , Dimerization , Growth Cones/drug effects , Humans , Ligands , Mice , Models, Molecular , Peptides/chemistry , Protein Structure, Quaternary , Protein Structure, Tertiary , Semaphorin-3A/metabolism , Semaphorin-3A/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship
14.
Mol Biol Cell ; 15(7): 3464-74, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15146055

ABSTRACT

Receptor tyrosine kinases have a single transmembrane (TM) segment that is usually assumed to play a passive role in ligand-induced dimerization and activation of the receptor. However, mutations within some of these receptors, and recent studies with the epidermal growth factor (EGF) and ErbB2 receptors have indicated that interactions between TM domains do contribute to stabilization of ligand-independent and/or ligand-induced receptor dimerization and activation. One consequence of the importance of these interactions is that short hydrophobic peptides corresponding to these domains should act as specific inhibitors. To test this hypothesis, we constructed expression vectors encoding short fusion peptides encompassing native or mutated TM domains of the EGF, ErbB2, and insulin receptors. In human cell lines overexpressing the wild-type EGF receptor or ErbB2, we observed that the peptides are expressed at the cell surface and that they inhibit specifically the autophosphorylation and signaling pathway of their cognate receptor. Identical results were obtained with peptides chemically synthesized. Mechanism of action involves inhibition of dimerization of the receptors as shown by the lack of effects of mutant nondimerizing sequences, completed by density centrifugation and covalent cross-linking experiments. Our findings stress the role of TM domain interactions in ErbB receptor function, and possibly for other single-spanning membrane proteins.


Subject(s)
Peptides/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Cell Surface/chemistry , Signal Transduction , Cell Line , Dimerization , ErbB Receptors/analysis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Peptides/chemistry , Peptides/genetics , Phosphorylation , Receptor, ErbB-2/genetics
15.
Crit Rev Oncol Hematol ; 50(1): 23-38, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15094157

ABSTRACT

Receptor tyrosine kinases (RTKs) are the main mediators of the signaling network that transmit extracellular signals into the cell, and control cellular differentiation and proliferation. Recent and rapid advances in our understanding of cellular signaling by receptor tyrosine kinases, in normal and malignant cells, have brought to light the potential of RTKs as selective anti-cancer targets. Their activity is normally tightly controlled and regulated. Overexpression of RTK proteins or functional alterations caused by mutations in the corresponding genes or abnormal stimulation by autocrine growth factor loops contribute to constitutive RTK signaling, resulting in dysregulated cell growth and cancer. The mechanisms of uncontrolled RTK signaling that leads to cancer has provided the rationale for anti-RTK drug development. Herceptin, Gleevec, and Iressa are the first examples of drugs which have successfully translated basic research on oncogenes into cancer therapeutics. RTKs can be viewed as multifunctional targets, and strategies towards the prevention and inhibition of RTK signaling include antibodies, antagonist ligands, small molecule inhibitors of protein kinase activity, and inhibitors of protein-protein interactions. Progresses in the field of rational drug design and computational chemistry will vastly benefit from the availability of increasing structural knowledge of both the kinase domains and the ligand-binding sites of these receptors.


Subject(s)
Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Drug Delivery Systems , Humans , Ligands , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...