Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 8531, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819167

ABSTRACT

For secreted proteins, proper protein folding is essential not only for biological function but also for secretion itself. Proteins with folding problems are trapped in the endoplasmic reticulum (ER) and are eventually degraded in the cytoplasm. In this study, we exploited co-expression of an artificial fusion protein, based on the sequence of a DnaJ protein, which could interact as co-chaperones in the Hsp70-based protein-folding system, with target recombinant secreted proteins to enhance their production and secretion. The J-domain sequence or a fragment thereof was conjugated to a target protein-binding domain that was capable of binding to a portion of the target-protein sequence. Production of many of the target proteins was significantly upregulated when co-expressed with the J-domain fusion protein. Surprisingly, the enhancement of secretion was observed even when the J-domain had a mutation in the HPD motif, which is necessary for J-protein-Hsp70 interactions, suggesting the phenomenon observed is independent on functional J-protein-Hsp70 interactions. This technology has great potential for not only enhancing the production of recombinant proteins, but also to treat conformational diseases such as cystic fibrosis, and Alpha-1 antitrypsin deficiency.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Protein Folding , Protein Transport , Recombinant Fusion Proteins/metabolism , HEK293 Cells , HSP40 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Protein Binding , Recombinant Fusion Proteins/genetics
2.
J Inorg Biochem ; 130: 69-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24176921

ABSTRACT

Elesclomol is a novel anticancer agent that has been evaluated in a number of late stage clinical trials. A new and convenient synthesis of elesclomol and its copper complex is described. X-ray crystallographic characterization and the electrochemical properties of the elesclomol copper(II) complex are discussed. The copper(II) cation is coordinated in a highly distorted square-planar geometry to each of the sulphur and amide nitrogen atoms of elesclomol. Electrochemical measurements demonstrate that the complex undergoes a reversible one-electron reduction at biologically accessible potentials. In contrast the free elesclomol is found electrochemically inactive. This evidence is in strong support of the mechanism of action we proposed for the anticancer activity of elesclomol.


Subject(s)
Copper/chemistry , Hydrazines/chemistry , Hydrazines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chemistry Techniques, Synthetic , Crystallography, X-Ray , Electrochemical Techniques , Hydrazines/chemical synthesis
3.
Toxicol Appl Pharmacol ; 273(2): 401-9, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24090817

ABSTRACT

In human trials certain heat shock protein 90 (Hsp90) inhibitors, including 17-DMAG and NVP-AUY922, have caused visual disorders indicative of retinal dysfunction; others such as 17-AAG and ganetespib have not. To understand these safety profile differences we evaluated histopathological changes and exposure profiles of four Hsp90 inhibitors, with or without clinical reports of adverse ocular effects, using a rat retinal model. Retinal morphology, Hsp70 expression (a surrogate marker of Hsp90 inhibition), apoptotic induction and pharmacokinetic drug exposure analysis were examined in rats treated with the ansamycins 17-DMAG and 17-AAG, or with the second-generation compounds NVP-AUY922 and ganetespib. Both 17-DMAG and NVP-AUY922 induced strong yet restricted retinal Hsp70 up-regulation and promoted marked photoreceptor cell death 24h after the final dose. In contrast, neither 17-AAG nor ganetespib elicited photoreceptor injury. When the relationship between drug distribution and photoreceptor degeneration was examined, 17-DMAG and NVP-AUY922 showed substantial retinal accumulation, with high retina/plasma (R/P) ratios and slow elimination rates, such that 51% of 17-DMAG and 65% of NVP-AUY922 present at 30 min post-injection were retained in the retina 6h post-dose. For 17-AAG and ganetespib, retinal elimination was rapid (90% and 70% of drugs eliminated from the retina at 6h, respectively) which correlated with lower R/P ratios. These findings indicate that prolonged inhibition of Hsp90 activity in the eye results in photoreceptor cell death. Moreover, the results suggest that the retina/plasma exposure ratio and retinal elimination rate profiles of Hsp90 inhibitors, irrespective of their chemical class, may predict for ocular toxicity potential.


Subject(s)
Disease Models, Animal , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Animals , Benzoquinones/toxicity , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/toxicity , Male , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Predictive Value of Tests , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Retinal Degeneration/pathology
4.
Bioorg Med Chem Lett ; 23(18): 5070-6, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23937981

ABSTRACT

A series of N'(1),N'(3)-dialkyl-N'(1),N'(3)-di(alkylcarbonothioyl) malonohydrazides have been designed and synthesized as anticancer agents by targeting oxidative stress and Hsp70 induction. Structure-activity relationship (SAR) studies lead to the discovery of STA-4783 (elesclomol), a novel small molecule that has been evaluated in a number of clinical trials as an anticancer agent in combination with Taxol.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Discovery , Hydrazines/pharmacology , Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Hydrazines/chemical synthesis , Hydrazines/chemistry , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Neoplasms, Experimental/pathology , Paclitaxel/chemistry , Paclitaxel/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
5.
Free Radic Biol Med ; 52(10): 2142-50, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22542443

ABSTRACT

Elesclomol is an investigational drug that exerts potent anticancer activity through the elevation of reactive oxygen species (ROS) levels and is currently under clinical evaluation as a novel anticancer therapeutic. Here we report the first description of selective mitochondrial ROS induction by elesclomol in cancer cells based on the unique physicochemical properties of the compound. Elesclomol preferentially chelates copper (Cu) outside of cells and enters as elesclomol-Cu(II). The elesclomol-Cu(II) complex then rapidly and selectively transports the copper to mitochondria. In this organelle Cu(II) is reduced to Cu(I), followed by subsequent ROS generation. Upon dissociation from the complex, elesclomol is effluxed from cells and repeats shuttling elesclomol-Cu complexes from the extracellular to the intracellular compartments, leading to continued copper accumulation within mitochondria. An optimal range of redox potentials exhibited by copper chelates of elesclomol and its analogs correlated with the elevation of mitochondrial Cu(I) levels and cytotoxic activity, suggesting that redox reduction of the copper triggers mitochondrial ROS induction. Importantly the mitochondrial selectivity exhibited by elesclomol is a distinct characteristic of the compound that is not shared by other chelators, including disulfiram. Together these findings highlight a unique mechanism of action with important implications for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/metabolism , Hydrazines/pharmacology , Mitochondria/metabolism , Neoplasms/drug therapy , Oxidative Stress/drug effects , Biological Transport/drug effects , Cell Line, Tumor , Copper/chemistry , Humans , Mitochondria/drug effects , Neoplasms/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism
6.
Invest New Drugs ; 30(6): 2201-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22227828

ABSTRACT

Systemic chemotherapy using two-drug platinum-based regimens for the treatment of advanced stage non-small cell lung cancer (NSCLC) has largely reached a plateau of effectiveness. Accordingly, efforts to improve survival and quality of life outcomes have more recently focused on the use of molecularly targeted agents, either alone or in combination with standard of care therapies such as taxanes. The molecular chaperone heat shock protein 90 (Hsp90) represents an attractive candidate for therapeutic intervention, as its inhibition results in the simultaneous blockade of multiple oncogenic signaling cascades. Ganetespib is a non-ansamycin inhibitor of Hsp90 currently under clinical evaluation in a number of human malignancies, including NSCLC. Here we show that ganetespib potentiates the cytotoxic activity of the taxanes paclitaxel and docetaxel in NSCLC models. The combination of ganetespib with paclitaxel, docetaxel or another microtubule-targeted agent vincristine resulted in synergistic antiproliferative effects in the H1975 cell line in vitro. These benefits translated to improved efficacy in H1975 xenografts in vivo, with significantly enhanced tumor growth inhibition observed in combination with paclitaxel and tumor regressions seen with docetaxel. Notably, concurrent exposure to ganetespib and docetaxel improved antitumor activity in 5 of 6 NSCLC xenograft models examined. Our data suggest that the improved therapeutic indices are likely to be mechanistically multifactorial, including loss of pro-survival signaling and direct cell cycle effects resulting from Hsp90 modulation by ganetespib. Taken together, these findings provide preclinical evidence for the use of this combination to treat patients with advanced NSCLC.


Subject(s)
Antineoplastic Agents/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Paclitaxel/administration & dosage , Taxoids/administration & dosage , Triazoles/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Drug Combinations , Female , Humans , Mice , Mice, SCID , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 11(2): 475-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22144665

ABSTRACT

Targeted inhibition of the molecular chaperone Hsp90 results in the simultaneous blockade of multiple oncogenic signaling pathways and has, thus, emerged as an attractive strategy for the development of novel cancer therapeutics. Ganetespib (formerly known as STA-9090) is a unique resorcinolic triazolone inhibitor of Hsp90 that is currently in clinical trials for a number of human cancers. In the present study, we showed that ganetespib exhibits potent in vitro cytotoxicity in a range of solid and hematologic tumor cell lines, including those that express mutated kinases that confer resistance to small-molecule tyrosine kinase inhibitors. Ganetespib treatment rapidly induced the degradation of known Hsp90 client proteins, displayed superior potency to the ansamycin inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG), and exhibited sustained activity even with short exposure times. In vivo, ganetespib showed potent antitumor efficacy in solid and hematologic xenograft models of oncogene addiction, as evidenced by significant growth inhibition and/or regressions. Notably, evaluation of the microregional activity of ganetespib in tumor xenografts showed that ganetespib was efficiently distributed throughout tumor tissue, including hypoxic regions >150 µm from the microvasculature, to inhibit proliferation and induce apoptosis. Importantly, ganetespib showed no evidence of cardiac or liver toxicity. Taken together, this preclinical activity profile indicates that ganetespib may have broad application for a variety of human malignancies, and with select mechanistic and safety advantages over other first- and second-generation Hsp90 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Triazoles/pharmacology , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Benzoquinones/adverse effects , Benzoquinones/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/etiology , Crystallography, X-Ray , Female , HL-60 Cells , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Heart/drug effects , Heart/physiology , Humans , K562 Cells , Lactams, Macrocyclic/adverse effects , Lactams, Macrocyclic/pharmacology , Male , Mice , Mice, Nude , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Rabbits , Rats , Rats, Sprague-Dawley , Triazoles/adverse effects , Triazoles/chemistry , Xenograft Model Antitumor Assays/methods
8.
PLoS One ; 6(4): e18552, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21533169

ABSTRACT

There is accumulating evidence that dysregulated JAK signaling occurs in a wide variety of cancer types. In particular, mutations in JAK2 can result in the constitutive activation of STAT transcription factors and lead to oncogenic growth. JAK kinases are established Hsp90 client proteins and here we show that the novel small molecule Hsp90 inhibitor ganetespib (formerly STA-9090) exhibits potent in vitro and in vivo activity in a range of solid and hematological tumor cells that are dependent on JAK2 activity for growth and survival. Of note, ganetespib treatment results in sustained depletion of JAK2, including the constitutively active JAK2(V617F) mutant, with subsequent loss of STAT activity and reduced STAT-target gene expression. In contrast, treatment with the pan-JAK inhibitor P6 results in only transient effects on these processes. Further differentiating these modes of intervention, RNA and protein expression studies show that ganetespib additionally modulates cell cycle regulatory proteins, while P6 does not. The concomitant impact of ganetespib on both cell growth and cell division signaling translates to potent antitumor efficacy in mouse models of xenografts and disseminated JAK/STAT-driven leukemia. Overall, our findings support Hsp90 inhibition as a novel therapeutic approach for combating diseases dependent on JAK/STAT signaling, with the multimodal action of ganetespib demonstrating advantages over JAK-specific inhibitors.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Janus Kinase 2/metabolism , Neoplasms, Experimental/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Triazoles/pharmacology , Animals , Cell Division/drug effects , Genes, cdc , Mice , Neoplasms, Experimental/pathology , Tumor Cells, Cultured
9.
Mov Disord ; 26(9): 1639-47, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21542017

ABSTRACT

Among heat shock proteins, mortalin has been linked to the pathogenesis of Parkinson's disease. In the present work a rat model of Parkinson's disease was used to analyze the expression of striatal proteins and, more specifically, mortalin expression. The possible involvement of mortalin in Parkinson's disease pathogenesis was further investigated by utilizing an electrophysiological approach and pharmacological inhibition of mortalin in both the physiological and the parkinsonian states. Proteomic analysis was used to investigate changes in striatal protein expression in the 6-hydroxydopamine rat model of Parkinson's disease. The electrophysiological effects of MKT-077, a rhodamine-123 analogue acting as an inhibitor of mortalin, were measured by field potential recordings from corticostriatal brain slices obtained from control, sham-operated, and 6-hydroxydopamine-denervated animals. Slices in the presence of rotenone, an inhibitor of mitochondrial complex I, were also analyzed. Proteomic analysis revealed downregulation of mortalin in the striata of 6-hydroxydopamine-treated rats in comparison with sham-operated animals. MKT-077 reduced corticostriatal field potential amplitude in physiological conditions, inducing membrane depolarization and inward current in striatal medium spiny neurons. In addition, we observed that concentrations of MKT-077 not inducing any electrophysiological effect in physiological conditions caused significant changes in striatal slices from parkinsonian animals as well as in slices treated with a submaximal concentration of rotenone. These findings suggest a critical link between mortalin function and mitochondrial activity in both physiological and pathological conditions mimicking Parkinson's disease.


Subject(s)
Antiparasitic Agents/therapeutic use , HSP70 Heat-Shock Proteins/therapeutic use , Parkinson Disease/drug therapy , Action Potentials/drug effects , Animals , Cerebral Cortex/pathology , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electron Transport Complex I/metabolism , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , In Vitro Techniques , Male , Neurons/drug effects , Oxidopamine/toxicity , Parkinson Disease/etiology , Parkinson Disease/pathology , Proteomics/methods , Pyridines/pharmacology , Rats , Rats, Wistar , Thiazoles/pharmacology
10.
Int J Cancer ; 126(6): 1428-35, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19739077

ABSTRACT

Mortalin, the mitochondrial hsp70, is a vital constitutively expressed heat shock protein. Its elevated expression has been correlated with malignant transformation and poor cancer prognosis. Cancer cells exhibit increased resistance to complement-dependent cytotoxicity, partly due to their capacity to eliminate the complement membrane attack complex (MAC) from their cell surface. As we have previously reported, mortalin and the complement membrane attack complexes are released in membrane vesicles from complement attacked cells. As shown here, knock down of mortalin with specific siRNA reduces MAC elimination and enhances cell sensitivity to MAC-induced cell death. Similar results were obtained with MKT-077, a cationic rhodacyanine dye that inhibits mortalin. Treatment of human erythroleukemia K562 and colorectal carcinoma HCT116 cells with MKT-077 sensitizes them to cell death mediated by MAC but not by streptolysin O. Pre-treatment of cells with MKT-077 also reduces the extent of MAC-mortalin vesiculation following a sublytic complement attack. In the presence of MKT-077, the direct binding of mortalin to complement C9, the major MAC component, is inhibited. The tumor suppressor protein p53 is a known mortalin client protein. The effect of MKT-077 on complement-mediated lysis of HCT116 p53(+/+) and p53(-/-) cells was found to be independent on the presence of p53. Our results also demonstrate that recombinant human mortain inhibits complement-mediated hemolysis of rabbit erythrocytes as well as zinc-induced C9 polymerization. We conclude that mortalin supports cancer cell resistance to complement-dependent cytotoxicity and propose consideration of mortalin as a novel target for cancer adjuvant immunotherapy.


Subject(s)
Complement C9/metabolism , HSP70 Heat-Shock Proteins/metabolism , RNA Interference , Animals , Bacterial Proteins/pharmacology , Blotting, Western , Calcimycin/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , HCT116 Cells , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , Hemolysis/drug effects , Humans , Ionophores/pharmacology , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Pyridines/pharmacology , Rabbits , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Streptolysins/pharmacology , Thiazoles/pharmacology
12.
Bioorg Med Chem Lett ; 16(19): 5164-8, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16870446

ABSTRACT

We report herein the SAR studies of a series of indole-imidazole compounds. that demonstrate substantial in vitro anti-proliferative activities against cancer cell lines, including multidrug resistance (MDR) phenotypes. The in vitro cytotoxic effects have been demonstrated across a wide array of tumor types, including hematologic and solid tumor cell lines of various origins (e.g., leukemia, breast, colon, and uterine).


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Resistance, Multiple , Imidazoles/chemical synthesis , Indoles/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inhibitory Concentration 50 , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...