Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Mol Pharm ; 19(5): 1422-1433, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35389227

ABSTRACT

With a wide range of available cytotoxic therapeutics, the main focus of current cancer research is to deliver them specifically to the cancer cells, minimizing toxicity against healthy tissues. Targeted therapy utilizes different carriers for cytotoxic drugs, combining a targeting molecule, typically an antibody, and a highly toxic payload. For the effective delivery of such cytotoxic conjugates, a molecular target on the cancer cell is required. Various proteins are exclusively or abundantly expressed in cancer cells, making them a possible target for drug carriers. Fibroblast growth factor receptor 1 (FGFR1) overexpression has been reported in different types of cancer, but no FGFR1-targeting cytotoxic conjugate has been approved for therapy so far. In this study, the FGFR1-targeting peptide previously described in the literature was reformatted into a peptibody-peptide fusion with the fragment crystallizable (Fc) domain of IgG1. PeptibodyC19 can be effectively internalized into FGFR1-overexpressing cells and does not induce cells' proliferation. The main challenge for its use as a cytotoxic conjugate is a cysteine residue located within the targeting peptide. A standard drug-conjugation strategy based on the maleimide-thiol reaction involves modification of cysteines within the Fc domain hinge region. Applied here, however, may easily result in the modification of the targeting peptide with the drug, limiting its affinity to the target and therefore the potential for specific drug delivery. To investigate if this is the case, we have performed conjugation reactions with different auristatin derivatives (PEGylated and unmodified) under various conditions. By controlling the reduction conditions and the type of cytotoxic payload, different numbers of cysteines were substituted, allowing us to avoid conjugating the drug to the targeting peptide, which could affect its binding to FGFR1. The optimized protocol with PEGylated auristatin yielded doubly substituted peptibodyC19, showing specific cytotoxicity toward the FGFR1-expressing lung cancer cells, with no effect on cells with low FGFR1 levels. Indeed, additional cysteine poses a risk of unwanted modification, but changes in the type of cytotoxic payload and reaction conditions allow the use of standard thiol-maleimide-based conjugation to achieve standard Fc hinge region cysteine modification, analogously to antibody-drug conjugates.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cysteine/chemistry , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Maleimides/chemistry , Polyethylene Glycols , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Sulfhydryl Compounds
2.
Front Pharmacol ; 12: 748936, 2021.
Article in English | MEDLINE | ID: mdl-34867353

ABSTRACT

Targeted therapies are a promising alternative to conventional chemotherapy, with an increasing number of therapeutics targeting specific molecular aberrancies in cancer cells. One of the emerging targets for directed cancer treatments is fibroblast growth factor receptors (FGFRs), which are known to be involved in the pathogenesis and progression of multiple cancer types, specially in lung, bladder, and breast cancers. Here, we are demonstrating the development of the FGFR1-targeting agent based on the interactome screening approach, based on the isolation of binding regions from ligands interacting with the receptor. The parallel analysis by FGFR1 pull-down of chymotryptic peptides coupled with MS analysis, and PepSpot analysis yielded equivalent peptide sequences from FGF4, one of the FGFR1 ligands. Three sequences served as a basis for peptibody (Fc-fusion) generation, to overcome clinical limitations of peptidic agents, and two of them showed favorable FGFR1-binding in vitro and FGFR1-dependent internalization into cells. To validate if developed FGFR1-targeting peptibodies can be used for drug delivery, similar to the well-established concept of antibody-drug conjugates (ADCs), peptibodyF4_1 was successfully conjugated with monomethylauristatin E (MMAE), and has shown significant and specific toxicity toward FGFR1-expressing lung cancer cell lines, with nanomolar EC50 values. Essentially, the development of new effective FGFR1 binders that comprise the naturally occurring FGFR-recognition peptides and Fc region ensuring high plasma stability, and long bloodstream circulation is an interesting strategy expanding targeted anticancer agents' portfolio. Furthermore, identifying peptides effectively binding the receptor from sequences of its ligands is not limited to FGFRs and is an approach versatile enough to be a basis for a new peptide/peptibodies development strategy.

3.
J Vis Exp ; (167)2021 01 05.
Article in English | MEDLINE | ID: mdl-33491672

ABSTRACT

Cancer is currently the second most common cause of death worldwide. The hallmark of cancer cells is the presence of specific marker proteins such as growth factor receptors on their surface. This feature enables development of highly selective therapeutics, the protein bioconjugates, composed of targeting proteins (antibodies or receptor ligands) connected to highly cytotoxic drugs by a specific linker. Due to very high affinity and selectivity of targeting proteins the bioconjugates recognize marker proteins on the cancer cells surface and utilize receptor-mediated endocytosis to reach the cell interior. Intracellular vesicular transport system ultimately delivers the bioconjugates to the lysosomes, where proteolysis separates free cytotoxic drugs from the proteinaceous core of the bioconjugates, triggering drug-dependent cancer cell death. Currently, there are several protein bioconjugates approved for cancer treatment and large number is under development or clinical trials. One of the main challenges in the generation of the bioconjugates is a site-specific attachment of the cytotoxic drug to the targeting protein. Recent years have brought a tremendous progress in the development of chemical and enzymatic strategies for protein modification with cytotoxic drugs. Here we present the detailed protocols for the site-specific incorporation of cytotoxic warheads into targeting proteins using a chemical method employing maleimide-thiol chemistry and an enzymatic approach that relies on sortase A-mediated ligation. We use engineered variant of fibroblast growth factor 2 and fragment crystallizable region of human immunoglobulin G as an exemplary targeting proteins and monomethyl auristatin E and methotrexate as model cytotoxic drugs. All the described strategies allow for highly efficient generation of biologically active cytotoxic conjugates of defined molecular architecture with potential for selective treatment of diverse cancers.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Maleimides/chemistry , Sulfhydryl Compounds/chemistry , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Fibroblast Growth Factor 2/metabolism , Humans , Immunoglobulin Fc Fragments/chemistry , Neoplasms/drug therapy , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protein Domains , Protein Engineering
4.
Cancers (Basel) ; 12(10)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076489

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are emerging targets for directed cancer therapy. Presented here is a new FGFR1-targeting conjugate, the peptibodyF2, which employs peptibody, a fusion of peptide and the Fc fragment of human IgG as a selective targeting agent and drug carrier. Short peptide based on FGF2 sequence was used to construct a FGFR1-targeting peptibody. We have shown that this peptide ensures specific delivery of peptibodyF2 into FGFR1-expressing cells. In order to use peptibodyF2 as a delivery vehicle for cytotoxic drugs, we have conjugated it with MMAE, a drug widely used in antibody-drug conjugates for targeted therapy. Resulting conjugate shows high and specific cytotoxicity towards FGFR1-positive cells, i.e., squamous cell lung carcinoma NCI-H520, while remaining non-toxic for FGFR1-negative cells. Such peptibody-drug conjugate can serve as a basis for development of therapy for tumors with overexpressed or malfunctioning FGFRs.

5.
Cells ; 8(8)2019 08 15.
Article in English | MEDLINE | ID: mdl-31443196

ABSTRACT

Fibroblast growth factor 1 (FGF1) has been shown to interact with integrin αvß3 through a specific binding site, involving Arg35 residue. The FGF1 mutant (R35E) with impaired integrin binding was found to be defective in its proliferative response, although it was still able to interact with FGF receptors (FGFR) and heparin and induce the activation of downstream signaling pathways. Here, we demonstrate that the lack of mitogenic potential of R35E mutant is directly caused by its decreased thermodynamic stability and susceptibility to proteolytic degradation. Introduction of three stabilizing mutations into R35E variant compensated the effect of destabilizing R35E mutation and restored the proliferation potential of FGF1. Moreover, the stabilized R35E variant regained both anti-apoptotic and wound healing activities, while remaining defective in binding to integrin αvß3. Our results suggest that the thermodynamic stability and resistance to degradation, rather than the interaction with integrin are required for mitogenic response of FGF1.


Subject(s)
Fibroblast Growth Factor 1/chemistry , Integrin alphaVbeta3/metabolism , Protein Stability , Proteolysis , Animals , Binding Sites , Fibroblast Growth Factor 1/genetics , Heparin/chemistry , Humans , Integrin alphaVbeta3/chemistry , Kinetics , Mice , Mutation , NIH 3T3 Cells , Protein Binding , Receptors, Fibroblast Growth Factor/chemistry
6.
FEBS Open Bio ; 9(5): 914-924, 2019 05.
Article in English | MEDLINE | ID: mdl-30968602

ABSTRACT

Overexpression of fibroblast growth factor receptor 1 (FGFR1) is a common aberration in lung and breast cancers and has necessitated the design of drugs targeting FGFR1-dependent downstream signaling and FGFR1 ligand binding. To date, the major group of drugs being developed for treatment of FGFR1-dependent cancers are small-molecule tyrosine kinase inhibitors; however, the limited specificity of these drugs has led to increasing attempts to design molecules targeting the extracellular domain of FGFR1. Here, we used the phage display technique to select cyclic peptides F8 (ACSLNHTVNC) and G10 (ACSAKTTSAC) as binders of the fibroblast growth factor 1 (FGF1)-FGFR1 interface. ELISA and in vitro cell assays were performed to reveal that cyclic peptide F8 is more effective in preventing the FGF1-FGFR1 interaction, and also decreases FGF1-induced proliferation of BA/F3 FGFR1c cells by over 40%. Such an effect was not observed for BA/F3 cells lacking FGFR1. Therefore, cyclic peptide F8 can act as a FGF1-FGFR1 interaction antagonist, and may be suitable for further development for potential use in therapies against FGFR1-expressing cancer cells.


Subject(s)
Bacteriophages/metabolism , Fibroblast Growth Factor 1/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Peptides, Cyclic/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Animals , Cell Line , Cell Surface Display Techniques , Gene Expression Profiling , Mice , NIH 3T3 Cells , Signal Transduction
7.
Int J Mol Sci ; 19(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134556

ABSTRACT

Fibroblast growth factor 1 (FGF1) and its receptors (FGFRs) regulate crucial biological processes such as cell proliferation and differentiation. Aberrant activation of FGFRs by their ligands can promote tumor growth and angiogenesis in many tumor types, including lung or breast cancer. The development of FGF1-targeting molecules with potential implications for the therapy of FGF1-driven tumors is recently being considered a promising approach in the treatment of cancer. In this study we have used phage display selection to find scFv antibody fragments selectively binding FGF1 and preventing it from binding to its receptor. Three identified scFv clones were expressed and characterized with regard to their binding to FGF1 and ability to interfere with FGF1-induced signaling cascades activation. In the next step the scFvs were cloned to scFv-Fc format, as dimeric Fc fusions prove beneficial in prospective therapeutic application. As expected, scFvs-Fc exhibited significantly increased affinity towards FGF1. We observed strong antiproliferative activity of the scFvs and scFvs-Fc in the in vitro cell models. Presented antibody fragments serve as novel FGF1 inhibitors and can be further utilized as powerful tools to use in the studies on the selective cancer therapy.


Subject(s)
Fibroblast Growth Factor 1/antagonists & inhibitors , Peptide Library , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clone Cells , Cloning, Molecular , Cross Reactions , Escherichia coli/genetics , Escherichia coli/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Mice , NIH 3T3 Cells , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Signal Transduction , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics
8.
Int J Mol Sci ; 19(7)2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30029518

ABSTRACT

In the rapidly developing field of targeted cancer therapy there is growing interest towards therapeutics combining two or more compounds to achieve synergistic action and minimize the chance of cancer resistance to treatment. We developed a fibroblast growth factor 2 (FGF2)-conjugate bearing two cytotoxic drugs with independent mode of action: α-amanitin and monomethyl auristatin E. Drugs are covalently attached to the targeting protein in a site-specific manner via maleimide-thiol conjugation and Cu(I)-catalyzed alkyne-azide cycloaddition. The dual warhead conjugate binds to FGF receptor 1 (FGFR1) and utilizes receptor-mediated endocytosis for selective internalization into cancer cells with FGFR1. The developed conjugate displays high cytotoxicity towards all tested FGFR1-positive cell lines. Most importantly, the improved cytotoxic effect of both drugs is observed for lung cancer cell line NCI-H446. The single drug-FGF2 conjugates have no impact on the viability of NCI-H446 cells, whereas the dual warhead-FGF2 conjugate selectively and efficiently kills these FGFR1 positive cancer cells. Due to the diversified mode of action the dual warhead-FGF2 conjugate may overcome the potential acquired resistance of FGFR1-overproducing cancer cells towards single cytotoxic drugs.


Subject(s)
Alpha-Amanitin/pharmacology , Fibroblast Growth Factor 2/metabolism , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Alpha-Amanitin/chemistry , Animals , Cell Line, Tumor , Endocytosis , Fibroblast Growth Factor 2/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Oligopeptides/chemistry , Protein Structure, Secondary , Signal Transduction
9.
Aging Cell ; 16(6): 1414-1424, 2017 12.
Article in English | MEDLINE | ID: mdl-29024389

ABSTRACT

Protein aggregation is enhanced upon exposure to various stress conditions and aging, which suggests that the quality control machinery regulating protein homeostasis could exhibit varied capacities in different stages of organismal lifespan. Recently, an efficient metazoan disaggregase activity was identified in vitro, which requires the Hsp70 chaperone and Hsp110 nucleotide exchange factor, together with single or cooperating J-protein co-chaperones of classes A and B. Here, we describe how the orthologous Hsp70s and J-protein of Caenorhabditis elegans work together to resolve protein aggregates both in vivo and in vitro to benefit organismal health. Using an RNAi knockdown approach, we show that class A and B J-proteins cooperate to form an interactive flexible network that relocalizes to protein aggregates upon heat shock and preferentially recruits constitutive Hsc70 to disaggregate heat-induced protein aggregates and polyQ aggregates that form in an age-dependent manner. Cooperation between class A and B J-proteins is also required for organismal health and promotes thermotolerance, maintenance of fecundity, and extended viability after heat stress. This disaggregase function of J-proteins and Hsc70 therefore constitutes a powerful regulatory network that is key to Hsc70-based protein quality control mechanisms in metazoa with a central role in the clearance of aggregates, stress recovery, and organismal fitness in aging.


Subject(s)
Heat-Shock Proteins/metabolism , Protein Aggregates/physiology , Aging , Animals , Caenorhabditis elegans/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Stress, Physiological
10.
Bioconjug Chem ; 28(7): 1850-1858, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28598150

ABSTRACT

Site-specific conjugation is a leading trend in the development of protein conjugates, including antibody-drug conjugates (ADCs), suitable for targeted cancer therapy. Here, we present a very efficient strategy for specific attachment of a cytotoxic drug to fibroblast growth factor 1 (FGF1), a natural ligand of FGF receptors (FGFRs), which are over-expressed in several types of lung, breast, and gastric cancers and are therefore an attractive molecular target. Recently, we showed that FGF1 fused to monomethylauristatin E (vcMMAE) was highly cytotoxic to cells presenting FGFRs on their surface and could be used as a targeting agent alternative to an antibody. Unfortunately, conjugation via maleimide chemistry to endogenous FGF1 cysteines or a cysteine introduced at the N-terminus proceeded with low yield and led to nonhomogeneous products. To improve the conjugation, we introduced a novel Lys-Cys-Lys motif at either FGF1 terminus, which increased cysteine reactivity and allowed us to obtain an FGF1 conjugate with a defined site of conjugation and a yield exceeding 95%. Using FGFR-expressing cancer lines, we confirmed specific cytotoxity of the obtained C-terminal FGF1-vcMMAE conjugate and its selective endocytososis as compared with FGFR1-negative cells. This simple and powerful approach relying on the introduction of a short sequence containing cysteine and positively charged amino acids could be used universally to improve the efficiency of the site-specific chemical modification of other proteins.


Subject(s)
Antineoplastic Agents/chemistry , Drug Delivery Systems/methods , Fibroblast Growth Factor 1/chemistry , Oligopeptides/chemistry , Amino Acid Motifs , Animals , Binding Sites , Cell Death/drug effects , Cell Line, Tumor , Cysteine/chemistry , Endocytosis/drug effects , Fibroblast Growth Factor 1/metabolism , Humans , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism
11.
Elife ; 62017 05 15.
Article in English | MEDLINE | ID: mdl-28504929

ABSTRACT

Hsp70 participates in a broad spectrum of protein folding processes extending from nascent chain folding to protein disaggregation. This versatility in function is achieved through a diverse family of J-protein cochaperones that select substrates for Hsp70. Substrate selection is further tuned by transient complexation between different classes of J-proteins, which expands the range of protein aggregates targeted by metazoan Hsp70 for disaggregation. We assessed the prevalence and evolutionary conservation of J-protein complexation and cooperation in disaggregation. We find the emergence of a eukaryote-specific signature for interclass complexation of canonical J-proteins. Consistently, complexes exist in yeast and human cells, but not in bacteria, and correlate with cooperative action in disaggregation in vitro. Signature alterations exclude some J-proteins from networking, which ensures correct J-protein pairing, functional network integrity and J-protein specialization. This fundamental change in J-protein biology during the prokaryote-to-eukaryote transition allows for increased fine-tuning and broadening of Hsp70 function in eukaryotes.


Subject(s)
Escherichia coli Proteins/chemistry , Evolution, Molecular , Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Protein Aggregates , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Models, Molecular , Molecular Chaperones/metabolism , Phylogeny , Protein Conformation , Protein Folding , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism
12.
J Mol Biol ; 428(21): 4378-4391, 2016 10 23.
Article in English | MEDLINE | ID: mdl-27616763

ABSTRACT

Escherichia coli ClpB and Saccharomyces cerevisiae Hsp104 are members of the Hsp100 family of ring-forming hexameric AAA+ chaperones that promote the solubilization of aggregated proteins and the propagation of prions. ClpB and Hsp104 cooperate with cognate Hsp70 chaperones for substrate targeting and activation of ATPase and substrate threading, achieved by transient Hsp70 binding to the repressing ClpB/Hsp104 M-domain. Fundamental differences in ATPase regulation and disaggregation mechanisms have been reported; however, these differences are raising doubts regarding the working principle of this AAA+ chaperone. In particular, unique functional plasticity was suggested to specifically enable Hsp104 to circumvent Hsp70 requirement for derepression in protein disaggregation and prion propagation. We show here that both ClpB and Hsp104 cooperation with Hsp70 is crucial for efficient protein disaggregation and, in contrast to earlier claims, cannot be circumvented by activating M-domain mutations. Activation of ClpB and Hsp104 requires two signals, relief of M-domain repression and substrate binding, leading to increased ATPase subunit coupling. These data demonstrate that ClpB and Hsp104 operate by the same basic mechanism, underscore a dominant function of Hsp70 in regulating ClpB/Hsp104 activity, and explain a plethora of in vivo studies showing a crucial function of Hsp70 in proteostasis and prion propagation.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Endopeptidase Clp , Protein Aggregates , Protein Interaction Mapping
13.
Drug Des Devel Ther ; 10: 2547-60, 2016.
Article in English | MEDLINE | ID: mdl-27563235

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are attractive candidate cancer therapy targets as they are overexpressed in multiple types of tumors, such as breast, prostate, bladder, and lung cancer. In this study, a natural ligand of FGFR, an engineered variant of fibroblast growth factor 1 (FGF1V), was conjugated to a potent cytotoxic drug, monomethyl auristatin E (MMAE), and used as a targeting agent for cancer cells overexpressing FGFRs, similar to antibodies in antibody-drug conjugates. The FGF1V-valine-citrulline-MMAE conjugate showed a favorable stability profile, bound FGFRs on the cell surface specifically, and efficiently released the drug (MMAE) upon cleavage by the lysosomal protease cathepsin B. Importantly, the conjugate showed a prominent cytotoxic effect toward cell lines expressing FGFR. FGF1V-vcMMAE was highly cytotoxic at concentrations even an order of magnitude lower than those found for free MMAE. This effect was FGFR-specific as cells lacking FGFR did not show any increased mortality.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Fibroblast Growth Factor 1/chemistry , Neoplasms/drug therapy , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/therapeutic use , Humans , Mice , Models, Molecular , Molecular Structure , NIH 3T3 Cells , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship
14.
Mol Cell ; 59(5): 781-93, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26300264

ABSTRACT

Intracellular amyloid fibrils linked to neurodegenerative disease typically accumulate in an age-related manner, suggesting inherent cellular capacity for counteracting amyloid formation in early life. Metazoan molecular chaperones assist native folding and block polymerization of amyloidogenic proteins, preempting amyloid fibril formation. Chaperone capacity for amyloid disassembly, however, is unclear. Here, we show that a specific combination of human Hsp70 disaggregase-associated chaperone components efficiently disassembles α-synuclein amyloid fibrils characteristic of Parkinson's disease in vitro. Specifically, the Hsc70 chaperone, the class B J-protein DNAJB1, and an Hsp110 family nucleotide exchange factor (NEF) provide ATP-dependent activity that disassembles amyloids within minutes via combined fibril fragmentation and depolymerization. This ultimately generates non-toxic α-synuclein monomers. Concerted, rapid interaction cycles of all three chaperone components with fibrils generate the power stroke required for disassembly. This identifies a powerful human Hsp70 disaggregase activity that efficiently disassembles amyloid fibrils and points to crucial yet undefined biology underlying amyloid-based diseases.


Subject(s)
Amyloid/metabolism , HSP70 Heat-Shock Proteins/metabolism , Parkinson Disease/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Electron Microscope Tomography , HSC70 Heat-Shock Proteins/metabolism , HSP110 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Humans , In Vitro Techniques , Kinetics , Molecular Chaperones/metabolism , Parkinson Disease/etiology , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Multimerization , Solubility , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism
15.
Nature ; 524(7564): 247-51, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26245380

ABSTRACT

Protein aggregates are the hallmark of stressed and ageing cells, and characterize several pathophysiological states. Healthy metazoan cells effectively eliminate intracellular protein aggregates, indicating that efficient disaggregation and/or degradation mechanisms exist. However, metazoans lack the key heat-shock protein disaggregase HSP100 of non-metazoan HSP70-dependent protein disaggregation systems, and the human HSP70 system alone, even with the crucial HSP110 nucleotide exchange factor, has poor disaggregation activity in vitro. This unresolved conundrum is central to protein quality control biology. Here we show that synergic cooperation between complexed J-protein co-chaperones of classes A and B unleashes highly efficient protein disaggregation activity in human and nematode HSP70 systems. Metazoan mixed-class J-protein complexes are transient, involve complementary charged regions conserved in the J-domains and carboxy-terminal domains of each J-protein class, and are flexible with respect to subunit composition. Complex formation allows J-proteins to initiate transient higher order chaperone structures involving HSP70 and interacting nucleotide exchange factors. A network of cooperative class A and B J-protein interactions therefore provides the metazoan HSP70 machinery with powerful, flexible, and finely regulatable disaggregase activity and a further level of regulation crucial for cellular protein quality control.


Subject(s)
Caenorhabditis elegans/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein Aggregates , Animals , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Humans , Models, Molecular , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control , Protein Binding , Protein Structure, Tertiary , Static Electricity
16.
Int J Nanomedicine ; 7: 5915-27, 2012.
Article in English | MEDLINE | ID: mdl-23226697

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are overexpressed in a wide variety of tumors, such as breast, bladder, and prostate cancer, and therefore they are attractive targets for different types of anticancer therapies. In this study, we designed, constructed, and characterized FGFR-targeted gold nanoconjugates suitable for infrared-induced thermal ablation (localized heating leading to cancer cell death) based on gold nanoparticles (AuNPs). We showed that a recombinant ligand of all FGFRs, human fibroblast growth factor 1 (FGF1), can be used as an agent targeting covalently bound AuNPs to cancer cells overexpressing FGFRs. To assure thermal stability, protease resistance, and prolonged half-life of the targeting protein, we employed highly stable FGF1 variant that retains the biological activities of the wild type FGF1. Novel FGF1 variant, AuNP conjugates are specifically internalized only by the cells expressing FGFRs, and they significantly reduce their viability after irradiation with near-infrared light (down to 40% of control cell viability), whereas the proliferation potential of cells lacking FGFRs is not affected. These results demonstrate the feasibility of FGF1-coated AuNPs for targeted cancer therapy.


Subject(s)
Fibroblast Growth Factor 1/pharmacokinetics , Gold/therapeutic use , Hyperthermia, Induced/methods , Metal Nanoparticles/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Cell Line, Tumor , Cell Survival/radiation effects , Gold/chemistry , Humans , Infrared Rays/therapeutic use , Metal Nanoparticles/chemistry , Treatment Outcome
17.
Curr Pharm Biotechnol ; 12(11): 1792-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21902634

ABSTRACT

Over the last two decades proteins have become increasingly important in human therapy and diagnosis. Engineering therapeutic proteins through improving their biological activity and stability has been a major interest in our group. In this mini-review we summarize our research on three proteins with pharmaceutical potential - serine protease inhibitor from squash seeds (CMTI), bovine pancreatic trypsin inhibitor (BPTI), and human fibroblast growth factor 1 (FGF1). To improve the functional properties of these proteins we used multiple techniques such as homology approach, rational design, total chemical synthesis, site-directed mutagenesis and phage display. The physicochemical properties of the obtained protein variants were evaluated using protein crystallography, spectroscopic techniques, enzymatic assays, stability measurements as well as numerous biological tests.


Subject(s)
Aprotinin , Drug Discovery/methods , Fibroblast Growth Factor 1 , Protein Engineering/methods , Serine Proteinase Inhibitors , Animals , Aprotinin/chemistry , Aprotinin/genetics , Aprotinin/pharmacology , Cattle , Cucurbita/chemistry , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/pharmacology , Humans , Mutagenesis, Site-Directed , Protein Stability , Seeds/chemistry , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/pharmacology
18.
Biotechnol Adv ; 29(4): 436-41, 2011.
Article in English | MEDLINE | ID: mdl-21443940

ABSTRACT

An increasing number of proteins are currently available on the market as therapeutics and this branch of the pharmaceutical industry will expand substantially during the coming years. As many diseases result from dysfunction of proteins forming multicomponent complexes, protein drugs with their inherent high specificity and affinity seem to be optimal medical agents. On the other hand, proteins are often highly instable and sensitive to degradation, which questions their applicability as effective therapeutics. Therefore, redesign and engineering of proteins is usually a required step in the present day drug development. Several approaches have been applied to optimize the protein properties central to their pharmaceutical use. This review focuses on different strategies that improve two crucial factors influencing protein drug efficiency: protein stability and its in vivo half-life. We provide examples of successful genetic and chemical modifications applied in the design of effective protein therapeutics.


Subject(s)
Pharmaceutical Preparations , Protein Engineering/methods , Proteins/therapeutic use , Genetic Engineering , Humans , Recombinant Fusion Proteins/therapeutic use
19.
Acta Crystallogr C ; 66(Pt 3): o119-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20203407

ABSTRACT

Comparison of the crystal structures of two pentadehydropeptides containing DeltaPhe residues, namely (Z,Z)-N-(tert-butoxycarbonyl)glycyl-alpha,beta-phenylalanylglycyl-alpha,beta-phenylalanylglycine (or Boc(0)-Gly(1)-Delta(Z)Phe(2)-Gly(3)-Delta(Z)Phe(4)-Gly(5)-OH) methanol solvate, C(29)H(33)N(5)O(8) x CH(4)O, (I), and (E,E)-N-(tert-butoxycarbonyl)glycyl-alpha,beta-phenylalanylglycyl-alpha,beta-phenylalanylglycine (or Boc(0)-Gly(1)-Delta(E)Phe(2)-Gly(3)-Delta(E)Phe(4)-Gly(5)-OH), C(29)H(33)N(5)O(8), (II), indicates that the Delta(Z)Phe residue is a more effective inducer of folded structures than the Delta(E)Phe residue. The values of the torsion angles phi and psi show the presence of two type-III' beta-turns at the Delta(Z)Phe residues and one type-II beta-turn at the Delta(E)Phe residue. All amino acids are linked trans to each other in both peptides. Beta-turns present in the peptides are stabilized by intramolecular 4-->1 hydrogen bonds. Molecules in both structures form two-dimensional hydrogen-bond networks parallel to the (100) plane.


Subject(s)
Oligopeptides/chemistry , Phenylalanine/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Hydrogen Bonding , Molecular Sequence Data , Molecular Structure , Protein Binding , Protein Conformation
20.
Nature ; 461(7262): 361-6, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19675567

ABSTRACT

Targeting of newly synthesized membrane proteins to the endoplasmic reticulum is an essential cellular process. Most membrane proteins are recognized and targeted co-translationally by the signal recognition particle. However, nearly 5% of membrane proteins are 'tail-anchored' by a single carboxy-terminal transmembrane domain that cannot access the co-translational pathway. Instead, tail-anchored proteins are targeted post-translationally by a conserved ATPase termed Get3. The mechanistic basis for tail-anchored protein recognition or targeting by Get3 is not known. Here we present crystal structures of yeast Get3 in 'open' (nucleotide-free) and 'closed' (ADP.AlF(4)(-)-bound) dimer states. In the closed state, the dimer interface of Get3 contains an enormous hydrophobic groove implicated by mutational analyses in tail-anchored protein binding. In the open state, Get3 undergoes a striking rearrangement that disrupts the groove and shields its hydrophobic surfaces. These data provide a molecular mechanism for nucleotide-regulated binding and release of tail-anchored proteins during their membrane targeting by Get3.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Aluminum Compounds/chemistry , Aluminum Compounds/metabolism , Crystallography, X-Ray , Fluorides/chemistry , Fluorides/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/chemistry , Methanococcus , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , SEC Translocation Channels , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...