Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Neurotherapeutics ; 21(1): e00291, 2024 Jan.
Article En | MEDLINE | ID: mdl-38241154

Alzheimer's disease (AD) is the leading cause of dementia and lacks highly effective treatments. Tau-based therapies hold promise. Tau reduction prevents amyloid-ß-induced dysfunction in preclinical models of AD and also prevents amyloid-ß-independent dysfunction in diverse disease models, especially those with network hyperexcitability, suggesting that strategies exploiting the mechanisms underlying Tau reduction may extend beyond AD. Tau binds several SH3 domain-containing proteins implicated in AD via its central proline-rich domain. We previously used a peptide inhibitor to demonstrate that blocking Tau interactions with SH3 domain-containing proteins ameliorates amyloid-ß-induced dysfunction. Here, we identify a top hit from high-throughput screening for small molecules that inhibit Tau-FynSH3 interactions and describe its optimization with medicinal chemistry. The resulting lead compound is a potent cell-permeable Tau-SH3 interaction inhibitor that binds Tau and prevents amyloid-ß-induced dysfunction, including network hyperexcitability. These data support the potential of using small molecule Tau-SH3 interaction inhibitors as a novel therapeutic approach to AD.


Alzheimer Disease , tau Proteins , Humans , tau Proteins/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , High-Throughput Screening Assays
2.
Antioxidants (Basel) ; 11(10)2022 Sep 23.
Article En | MEDLINE | ID: mdl-36290611

Acute kidney injury (AKI) is a major public health concern with significant morbidity and mortality and no current treatments beyond supportive care and dialysis. Preclinical studies have suggested that heme-oxygenase-1 (HO-1), an enzyme that catalyzes the breakdown of heme, has promise as a potential therapeutic target for AKI. Clinical trials involving HO-1 products (biliverdin, carbon monoxide, and iron), however, have not progressed beyond the Phase ½ level. We identified small-molecule inducers of HO-1 that enable us to exploit the full therapeutic potential of HO-1, the combination of its products, and yet-undefined effects of the enzyme system. Through cell-based, high-throughput screens for induction of HO-1 driven by the human HO-1 promoter/enhancer, we identified two novel small molecules and broxaldine (an FDA-approved drug) for further consideration as candidate compounds exhibiting an Emax ≥70% of 5 µM hemin and EC50 <10 µM. RNA sequencing identified shared binding motifs to NRF2, a transcription factor known to regulate antioxidant genes, including HMOX1. In vitro, the cytoprotective function of the candidates was assessed against cisplatin-induced cytotoxicity and apoptosis. In vivo, delivery of a candidate compound induced HO-1 expression in the kidneys of mice. This study serves as the basis for further development of small-molecule HO-1 inducers as preventative or therapeutic interventions for a variety of pathologies, including AKI.

3.
Bioorg Med Chem Lett ; 64: 128696, 2022 05 15.
Article En | MEDLINE | ID: mdl-35318165

Arsenicals belong to the class of chemical warfare agents known as vesicants, which are highly reactive, toxic and cause robust inflammatory response. Cutaneous exposure to arsenicals causes a wide range of systemic organ damage, beginning with cutaneous injuries, and later manifest multi-organ damage and death. Thus, the development of suitable antidotes that can effectively block injury following exposure to these agents is of great importance. Bromodomain 4 (BRD4), a member of the bromodomain and extra terminal domain (BET) family, plays crucial role in regulating transcription of inflammatory, proliferation and cell cycle genes. In this context, the development of potent small molecule inhibitors of BRD4 could serve as potential antidotes for arsenicals. Herein, we describe the synthesis and biological evaluation of a series of compounds.


Arsenicals , Anti-Inflammatory Agents/chemistry , Antidotes/pharmacology , Arsenicals/pharmacology , Arsenicals/therapeutic use , Nuclear Proteins/metabolism , Transcription Factors/metabolism
4.
Mol Cancer Ther ; 21(2): 347-358, 2022 02.
Article En | MEDLINE | ID: mdl-34907087

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM). The proteasome inhibitor bortezomib is one of the most effective first-line chemotherapeutic drugs for multiple myeloma; however, 15% to 20% of high-risk patients do not respond to or become resistant to this drug and the mechanisms of chemoresistance remain unclear. We previously demonstrated that multiple myeloma cells inhibit Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB), and that this OB-Runx2 deficiency induces a cytokine-rich and immunosuppressive microenvironment in the BM. In the current study, we assessed the impact of OB-Runx2 deficiency on the outcome of bortezomib treatment using OB-Runx2+/+ and OB-Runx2-/- mouse models of multiple myeloma. In vitro and in vivo experiments revealed that OB-Runx2 deficiency induces multiple myeloma cell resistance to bortezomib via the upregulation of immunosuppressive myeloid-derived suppressor cells (MDSCs), downregulation of cytotoxic T cells, and activation of TGFß1 in the BM. In multiple myeloma tumor-bearing OB-Runx2-/- mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)-mediated TGFß1 activation, reversed the BM immunosuppression and significantly reduced tumor burden. Furthermore, treatment with SRI31277 combined with bortezomib alleviated multiple myeloma cell resistance to bortezomib-induced apoptosis caused by OB-Runx2 deficiency in cocultured cells and produced a synergistic effect on tumor burden in OB-Runx2-/- mice. Depletion of MDSCs by 5-fluorouracil or gemcitabine similarly reversed the immunosuppressive effects and bortezomib resistance induced by OB-Runx2 deficiency in tumor-bearing mice, indicating the importance of the immune environment for drug resistance and suggesting new strategies to overcome bortezomib resistance in the treatment of multiple myeloma.


Bone Marrow/metabolism , Bortezomib/therapeutic use , Core Binding Factor Alpha 1 Subunit/deficiency , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Osteoblasts/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Bortezomib/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Multiple Myeloma/pathology
5.
Nat Commun ; 12(1): 4358, 2021 07 16.
Article En | MEDLINE | ID: mdl-34272367

Premature termination codons (PTCs) prevent translation of a full-length protein and trigger nonsense-mediated mRNA decay (NMD). Nonsense suppression (also termed readthrough) therapy restores protein function by selectively suppressing translation termination at PTCs. Poor efficacy of current readthrough agents prompted us to search for better compounds. An NMD-sensitive NanoLuc readthrough reporter was used to screen 771,345 compounds. Among the 180 compounds identified with readthrough activity, SRI-37240 and its more potent derivative SRI-41315, induce a prolonged pause at stop codons and suppress PTCs associated with cystic fibrosis in immortalized and primary human bronchial epithelial cells, restoring CFTR expression and function. SRI-41315 suppresses PTCs by reducing the abundance of the termination factor eRF1. SRI-41315 also potentiates aminoglycoside-mediated readthrough, leading to synergistic increases in CFTR activity. Combining readthrough agents that target distinct components of the translation machinery is a promising treatment strategy for diseases caused by PTCs.


Codon, Nonsense/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/drug effects , Nonsense Mediated mRNA Decay , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/metabolism , Aminoglycosides/metabolism , Codon, Nonsense/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Genes, Reporter , Gentamicins/pharmacology , HEK293 Cells , Humans , Microsomes, Liver/drug effects , Peptide Termination Factors/genetics , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Ribosomes/metabolism , Structure-Activity Relationship
6.
Antimicrob Agents Chemother ; 65(9): e0024421, 2021 08 17.
Article En | MEDLINE | ID: mdl-34152810

Venezuelan equine encephalitis virus (VEEV) is a reemerging alphavirus that can cause encephalitis resulting in severe human morbidity and mortality. Using a high-throughput cell-based screen, we identified a quinolinone compound that protected against VEEV-induced cytopathic effects. Analysis of viral replication in cells identified several quinolinone compounds with potent inhibitory activity against vaccine and virulent strains of VEEV. These quinolinones also displayed inhibitory activity against additional alphaviruses, such as Mayaro virus and Ross River virus, although the potency was greatly reduced. Time-of-addition studies indicated that these compounds inhibit the early-to-mid stage of viral replication. Deep sequencing and reverse genetics studies identified two unique resistance mutations in the nsP2 gene (Y102S/C; stalk domain) that conferred VEEV resistance on this chemical series. Moreover, introduction of a K102Y mutation into the nsP2 gene enhanced the sensitivity of chikungunya virus (CHIKV) to this chemical series. Computational modeling of CHIKV and VEEV nsP2 identified a highly probable docking alignment for the quinolinone compounds that require a tyrosine residue at position 102 within the helicase stalk domain. These studies identified a class of compounds with antiviral activity against VEEV and other alphaviruses and provide further evidence that therapeutics targeting nsP2 may be useful against alphavirus infection.


Chikungunya virus , Encephalitis Virus, Venezuelan Equine , Quinolones , Animals , Antiviral Agents/pharmacology , Encephalitis Virus, Venezuelan Equine/genetics , Horses , Humans , Quinolones/pharmacology , Virus Replication
7.
J Med Chem ; 64(8): 4762-4786, 2021 04 22.
Article En | MEDLINE | ID: mdl-33835811

A benzo[6]annulene, 4-(tert-butyl)-N-(3-methoxy-5,6,7,8-tetrahydronaphthalen-2-yl) benzamide (1a), was identified as an inhibitor against Chikungunya virus (CHIKV) with antiviral activity EC90 = 1.45 µM and viral titer reduction (VTR) of 2.5 log at 10 µM with no observed cytotoxicity (CC50 = 169 µM) in normal human dermal fibroblast cells. Chemistry efforts to improve potency, efficacy, and drug-like properties of 1a resulted in a novel lead compound 8q, which possessed excellent cellular antiviral activity (EC90 = 270 nM and VTR of 4.5 log at 10 µM) and improved liver microsomal stability. CHIKV resistance to an analog of 1a, compound 1c, tracked to a mutation in the nsP3 macrodomain. Further mechanism of action studies showed compounds working through inhibition of human dihydroorotate dehydrogenase in addition to CHIKV nsP3 macrodomain. Moderate efficacy was observed in an in vivo CHIKV challenge mouse model for compound 8q as viral replication was rescued from the pyrimidine salvage pathway.


Antiviral Agents/pharmacology , Benzene Derivatives/chemistry , Chikungunya virus/physiology , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Binding Sites , Cell Line , Cell Survival/drug effects , Chikungunya Fever/drug therapy , Dihydroorotate Dehydrogenase , Disease Models, Animal , Female , Half-Life , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Structure-Activity Relationship
8.
Eur J Med Chem ; 210: 112952, 2021 Jan 15.
Article En | MEDLINE | ID: mdl-33139114

ALS is a rare type of progressive neurological disease with unknown etiology. It results in the gradual degeneration and death of motor neurons responsible for controlling the voluntary muscles. Identification of mutations in the superoxide dismutase (SOD) 1 gene has been the most significant finding in ALS research. SOD1 abnormalities have been associated with both familial as well as sporadic ALS cases. SOD2 is a highly inducible SOD that performs in concurrence with SOD1 to detoxify ROS. Induction of SOD2 can be obtained through activation of NF-Ò¡Bs. We previously reported that SRI-22819 increases NF-Ò¡B expression and activation in vitro, but it has poor ADME properties in general and has no oral bioavailability. Our initial studies were focused on direct modifications of SRI-22819. There were active compounds identified but no improvement in microsomal stability was observed. In this context, we focused on making more significant structural changes in the core of the molecule. Ataluren, an oxadiazole compound that promotes read-through and expression of dystrophin in patients with Duchenne muscular dystrophy, bears some structural similarity to SRI-22819. Thus, we synthesized a series of SRI-22819 and Ataluren (PTC124) hybrid compounds. Several compounds from this series exhibited improved activity, microsomal stability and lower calculated polar surface area (PSA). This manuscript describes the synthesis and biological evaluation of SRI-22819 analogs and its hybrid combination with Ataluren.


Amyotrophic Lateral Sclerosis/drug therapy , NF-kappa B/agonists , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cell Line , Humans , Mice , Molecular Docking Simulation , NF-kappa B/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Structure-Activity Relationship , Superoxide Dismutase/metabolism
9.
Adv Virol ; 2020: 8844061, 2020.
Article En | MEDLINE | ID: mdl-33110426

Approximately 257 million people chronically infected with hepatitis B virus (HBV) worldwide are at risk of developing hepatocellular carcinoma (HCC). However, despite the availability of potent nucleoside/tide inhibitors, currently there are no curative therapies for chronic HBV infections. To identify potential new antiviral molecules, a select group of compounds previously evaluated in clinical studies were tested against 12 different viruses. Amongst the compounds tested, SRI-32007 (CYT997) demonstrated antiviral activity against HBV (genotype D) in HepG2.2.2.15 cell-based virus yield assay with 50% effective concentration (EC50) and selectivity index (SI) of 60.1 nM and 7.2, respectively. Anti-HBV activity of SRI-32007 was further confirmed against HBV genotype B in huh7 cells with secreted HBe antigen endpoint (EC50 40 nM and SI 250). To determine the stage of HBV life cycle inhibited by SRI-32007, time of addition experiment was conducted in HepG2-NTCP cell-based HBV infectious assay. Results indicated that SRI-32007 retained anti-HBV activity even when added 72 hours postinfection (72 h). Additional mechanism of action studies demonstrated potent inhibition of HBV core promoter activity by SRI-32007 with an EC50 of 40 nM and SI of >250. This study demonstrates anti-HBV activity of a repurposed compound SRI-32007 through inhibition of HBV core promoter activity. Further evaluation of SRI-32007 in HBV animal models is needed to confirm its activity in vivo. Our experiments illustrate the utility of repurposing strategy to identify novel antiviral chemical leads. HBV core promoter inhibitors such as SRI-32007 might enable the development of novel therapeutic strategies to combat HBV infections.

10.
Cell Metab ; 32(3): 353-365.e8, 2020 09 01.
Article En | MEDLINE | ID: mdl-32726606

Diabetes is characterized by hyperglycemia, loss of functional islet beta cell mass, deficiency of glucose-lowering insulin, and persistent alpha cell secretion of gluconeogenic glucagon. Still, no therapies that target these underlying processes are available. We therefore performed high-throughput screening of 300,000 compounds and extensive medicinal chemistry optimization and here report the discovery of SRI-37330, an orally bioavailable, non-toxic small molecule, which effectively rescued mice from streptozotocin- and obesity-induced (db/db) diabetes. Interestingly, in rat cells and in mouse and human islets, SRI-37330 inhibited expression and signaling of thioredoxin-interacting protein, which we have previously found to be elevated in diabetes and to have detrimental effects on islet function. In addition, SRI-37330 treatment inhibited glucagon secretion and function, reduced hepatic glucose production, and reversed hepatic steatosis. Thus, these studies describe a newly designed chemical compound that, compared to currently available therapies, may provide a distinct and effective approach to treating diabetes.


Carrier Proteins/genetics , Diabetes Mellitus, Experimental/drug therapy , Glucagon/metabolism , Hypoglycemic Agents/pharmacology , Small Molecule Libraries/pharmacology , Administration, Oral , Animals , Carrier Proteins/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Rats , Small Molecule Libraries/administration & dosage , Streptozocin
11.
ACS Med Chem Lett ; 11(6): 1130-1136, 2020 Jun 11.
Article En | MEDLINE | ID: mdl-32550992

TGF-ß has been a target of interest for the treatment of fibrotic diseases and certain cancers. Approaches to target TGF-ß include antagonists of the active ligand or TGF-ß receptor kinase activity. These approaches have failed in clinical trials due to a lack of effectiveness and a limited therapeutic window. In this context, newer and more selective approaches to target TGF-ß are needed. We previously reported that the matricellular protein, thrombospondin 1, activates the latent TGF-ß complex and that antagonism of this pathway using tri/tetrapeptides in various animal models reduces fibrosis. The tripeptide, SRI-31277 (1), is effective in vivo but has a short plasma half life (0.2 h). Herein we describe the design and synthesis SRI-31277 analogs, specifically smaller peptides that retain potency and have improved bioavailability. We identified SRI-35241 (36) with a single chiral center, which blocks TGF-ß activation (pIC50 = 8.12 nM) and has a plasma half life of 1.8 h (iv).

12.
Front Oncol ; 10: 241, 2020.
Article En | MEDLINE | ID: mdl-32185127

Resistance to radiation and chemotherapy in colorectal cancer (CRC) patients contribute significantly to refractory disease and disease progression. Herein, we provide mechanistic rationale for acquired or inherent chemotherapeutic resistance to the anti-tumor effects of 5-fluorouracil (5-FU) that is linked to oncogenic GLI1 transcription activity and NBS1 overexpression. Patients with high levels of GLI1 also expressed high levels of NBS1. Non-canonical activation of GLI1 is driven through oncogenic pathways in CRC, like the BRAFV600E mutation. GLI1 was identified as a novel regulator of NBS1 and discovered that by knocking down GLI1 levels in vitro, diminished NBS1 expression, increased DNA damage/apoptosis, and re-sensitization of 5-FU resistant cancer to treatment was observed. Furthermore, a novel GLI1 inhibitor, SRI-38832, which exhibited pharmacokinetic properties suitable for in vivo testing, was identified. GLI1 inhibition in a murine BRAFV600E variant xenograft model of CRC resulted in the same down-regulation of NBS1 observed in vitro as well as significant reduction of tumor growth/burden. GLI1 inhibition could therefore be a therapeutic option for 5-FU resistant and BRAFV600E variant CRC patients.

13.
Bioorg Med Chem Lett ; 30(4): 126950, 2020 02 15.
Article En | MEDLINE | ID: mdl-31928838

Amyotrophic lateral sclerosis (ALS) is a rare and progressive neurodegenerative disease with unknown etiology. It is caused by the degeneration of motor neurons responsible for controlling voluntary muscles. It has been reported that mutations in the superoxide dismutase (SOD) 1 gene can lead to ALS. SOD1 abnormalities have been identified in both familial, as well as sporadic ALS cases. SOD2 is a highly inducible SOD that works in conjunction with SOD1. SOD2 can be induced through activation of NF-κBs. We previously reported that the novel small molecule, SRI-22818, increases NF-κB expression and activation and SOD2 levels in vitro and has activity in vivo in the SOD1-G93A reference model of ALS. We report herein the synthesis and biological evaluation of SRI-22818 analogs.


Amyotrophic Lateral Sclerosis/pathology , Small Molecule Libraries/chemistry , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Gene Expression Regulation/drug effects , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
14.
ACS Infect Dis ; 5(12): 2014-2028, 2019 12 13.
Article En | MEDLINE | ID: mdl-31257853

Alphaviruses are arthropod-transmitted members of the Togaviridae family that can cause severe disease in humans, including debilitating arthralgia and severe neurological complications. Currently, there are no approved vaccines or antiviral therapies directed against the alphaviruses, and care is limited to treating disease symptoms. A phenotypic cell-based high-throughput screen was performed to identify small molecules that inhibit the replication of Venezuelan Equine Encephalitis Virus (VEEV). The compound, 1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-N-(3-fluoro-4-methoxybenzyl)ethan-1-amine (1), was identified as a highly active, potent inhibitor of VEEV with an effective concentration for 90% inhibition of virus (EC90) of 0.89 µM and 7.49 log reduction in virus titers at 10 µM concentration. These data suggest that further investigation of compound 1 as an antiviral therapeutic against VEEV, and perhaps other alphaviruses, is warranted. Experiments suggested that the antiviral activity of compound 1 is directed at an early step in the VEEV replication cycle by blocking viral RNA and protein synthesis.


Antiviral Agents/pharmacology , Benzylamines/pharmacology , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalomyelitis, Venezuelan Equine/virology , Animals , Antiviral Agents/chemistry , Benzylamines/chemistry , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Encephalomyelitis, Venezuelan Equine/drug therapy , High-Throughput Screening Assays , Humans , Molecular Structure , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects
15.
SLAS Discov ; 23(10): 1083-1091, 2018 12.
Article En | MEDLINE | ID: mdl-29958052

CC-chemokine receptor 7 (CCR7) is a G protein-coupled receptor expressed on a variety of immune cells. CCR7 plays a critical role in the migration of lymphocytes into secondary lymphoid tissues. CCR7 expression, however, has been linked to numerous disease states. Due to its therapeutic relevance and absence of available CCR7 inhibitors, we undertook a high-throughput screen (HTS) to identify small-molecule antagonists of the receptor. Here, we describe a robust HTS approach using a commercially available ß-galactosidase enzyme fragment complementation system and confirmatory transwell chemotaxis assays. This work resulted in the identification of several compounds with activity against CCR7. The most potent of these was subsequently determined to be cosalane, a cholesterol derivative previously designed as a therapeutic for human immunodeficiency virus. Cosalane inhibited both human and murine CCR7 in response to both CCL19 and CCL21 agonists at physiologic concentrations. Furthermore, cosalane produced durable inhibition of the receptor following a cellular incubation period with subsequent washout. Overall, our work describes the development of an HTS-compatible assay, completion of a large HTS campaign, and demonstration for the first time that cosalane is a validated CCR7 antagonist. These efforts could pave the way for new approaches to address CCR7-associated disease processes.


Aurintricarboxylic Acid/analogs & derivatives , High-Throughput Screening Assays , Receptors, CCR7/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Cell Line , Chemotaxis/drug effects , Drug Design , Humans , Ligands , Mice , Molecular Structure , Receptors, CCR7/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
17.
Matrix Biol ; 68-69: 28-43, 2018 08.
Article En | MEDLINE | ID: mdl-29288716

Transforming growth factor-ß (TGF-ß) is a central player in fibrotic disease. Clinical trials with global inhibitors of TGF-ß have been disappointing, suggesting that a more targeted approach is warranted. Conversion of the latent precursor to the biologically active form of TGF-ß represents a novel approach to selectively modulating TGF-ß in disease, as mechanisms employed to activate latent TGF-ß are typically cell, tissue, and/or disease specific. In this review, we will discuss the role of the matricellular protein, thrombospondin 1 (TSP-1), in regulation of latent TGF-ß activation and the use of an antagonist of TSP-1 mediated TGF-ß activation in a number of diverse fibrotic diseases. In particular, we will discuss the TSP-1/TGF-ß pathway in fibrotic complications of diabetes, liver fibrosis, and in multiple myeloma. We will also discuss emerging evidence for a role for TSP-1 in arterial remodeling, biomechanical modulation of TGF-ß activity, and in immune dysfunction. As TSP-1 expression is upregulated by factors induced in fibrotic disease, targeting the TSP-1/TGF-ß pathway potentially represents a more selective approach to controlling TGF-ß activity in disease.


Fibrosis/drug therapy , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Fibrosis/metabolism , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Multiple Myeloma/complications , Multiple Myeloma/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Thrombospondin 1/antagonists & inhibitors
18.
Oncotarget ; 8(53): 91459-91470, 2017 Oct 31.
Article En | MEDLINE | ID: mdl-29207657

Frizzled (Fzd) proteins are seven transmembrane receptors that belong to a novel and separated family of G-protein-coupled receptors (GPCRs). The Fzd receptors can respond to Wnt proteins to activate the canonical ß-catenin pathway which is important for both initiation and progression of cancers. Disruption of the Wnt/ß-catenin signal thus represents an opportunity for rational cancer prevention and therapy. Of the 10 members of the Fzd family, Fzd7 is the most important member involved in cancer development and progression. In the present studies, we applied structure-based virtual screening targeting the transmembrane domain (TMD) of Fzd7 to select compounds that could potentially bind to the Fzd7-TMD and block the Wnt/Fzd7 signaling and further evaluated them in biological assays. Six small molecule compounds were confirmed as Fzd7 inhibitors. The best hit, SRI37892, significantly blocked the Wnt/Fzd7 signaling with IC50 values in the sub-micromolar range and inhibited cancer cell proliferation with IC50 values around 2 µM. Our results provide the first proof of concept of targeting Fzd-TMD for the development of Wnt/Fzd modulators. The identified small molecular Fzd7 inhibitors can serve as a useful tool for studying the regulation mechanism(s) of Wnt/Fzd7 signaling as well as a starting point for the development of cancer therapeutic agents.

19.
Oncol Rep ; 38(5): 2814-2824, 2017 Nov.
Article En | MEDLINE | ID: mdl-29048650

Soft tissue sarcomas are rare and aggressive tumors arising from connective tissues. Adjuvant radiotherapy is a commonly used treatment approach for the majority of sarcomas. We attempted to identify a gene signature that can predict radiosensitive patients who are most likely to have a better treatment response from radiotherapy, compared with disease progression. Using the publicly available data of soft tissue sarcoma from The Cancer Genome Atlas, we developed a cross-validation procedure to identify a predictive gene signature for radiosensitivity. The results showed that the predicted radiosensitive patients who received radiotherapy had significantly improved treatment response. We further provide supportive evidence to validate our sensitivity prediction. Results showed that the predicted radiosensitive patients who received radiotherapy had significantly improved survival than patients who did not. ROC analysis showed that the developed gene signature had a powerful prediction on treatment response. We further found that predicted radiosensitive patients who received radiotherapy had a significantly reduced rate of new tumor events. Finally, we validated our gene signature using a hierarchical cluster analysis, and found that the predicted sensitivities were well-matched with results from the cluster analysis. These results are consistent with our expectation, suggesting that the identified gene signature and radiosensitivity prediction are effective. The genes involved in the signature may provide a molecular basis for prognostic studies and radiotherapy target discovery.


Computational Biology/methods , Gene Regulatory Networks/radiation effects , Sarcoma/radiotherapy , Sequence Analysis, RNA/methods , Aged , Cluster Analysis , Databases, Genetic , Disease Progression , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Middle Aged , Radiotherapy, Adjuvant , Sarcoma/genetics , Survival Analysis , Treatment Outcome
20.
Oncotarget ; 8(23): 37568-37583, 2017 Jun 06.
Article En | MEDLINE | ID: mdl-28455961

Patients with glioblastoma have one of the lowest overall survival rates among patients with cancer. Standard of care for patients with glioblastoma includes temozolomide and radiation therapy, yet 30% of patients do not respond to these treatments and nearly all glioblastoma tumors become resistant. Chlorpromazine is a United States Food and Drug Administration-approved phenothiazine widely used as a psychotropic in clinical practice. Recently, experimental evidence revealed the anti-proliferative activity of chlorpromazine against colon and brain tumors. Here, we used chemoresistant patient-derived glioma stem cells and chemoresistant human glioma cell lines to investigate the effects of chlorpromazine against chemoresistant glioma. Chlorpromazine selectively and significantly inhibited proliferation in chemoresistant glioma cells and glioma stem cells. Mechanistically, chlorpromazine inhibited cytochrome c oxidase (CcO, complex IV) activity from chemoresistant but not chemosensitive cells, without affecting other mitochondrial complexes. Notably, our previous studies revealed that the switch to chemoresistance in glioma cells is accompanied by a switch from the expression of CcO subunit 4 isoform 2 (COX4-2) to COX4-1. In this study, chlorpromazine induced cell cycle arrest selectively in glioma cells expressing COX4-1, and computer-simulated docking studies indicated that chlorpromazine binds more tightly to CcO expressing COX4-1 than to CcO expressing COX4-2. In orthotopic mouse brain tumor models, chlorpromazine treatment significantly increased the median overall survival of mice harboring chemoresistant tumors. These data indicate that chlorpromazine selectively inhibits the growth and proliferation of chemoresistant glioma cells expressing COX4-1. The feasibility of repositioning chlorpromazine for selectively treating chemoresistant glioma tumors should be further explored.


Brain Neoplasms/drug therapy , Chlorpromazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Electron Transport Complex IV/antagonists & inhibitors , Glioma/drug therapy , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antipsychotic Agents/pharmacology , Brain Neoplasms/metabolism , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dopamine Antagonists/pharmacology , Drug Repositioning , Electron Transport Complex IV/metabolism , Glioma/metabolism , Humans , Mice, Nude , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Temozolomide , Xenograft Model Antitumor Assays
...