119 results on '"Mark J. Suto"'
Search Results
2. Identification of Cytoprotective Small-Molecule Inducers of Heme-Oxygenase-1
- Author
-
Gelare Ghajar-Rahimi, Amie M. Traylor, Bini Mathew, James R. Bostwick, N Miranda Nebane, Anna A. Zmijewska, Stephanie K. Esman, Saakshi Thukral, Ling Zhai, Vijaya Sambandam, Rita M. Cowell, Mark J. Suto, James F. George, Corinne E. Augelli-Szafran, and Anupam Agarwal
- Subjects
acute kidney injury ,heme oxygenase ,cisplatin nephrotoxicity ,small-molecule drugs ,high-throughput screen ,Therapeutics. Pharmacology ,RM1-950 - Abstract
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 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.
- Published
- 2022
- Full Text
- View/download PDF
3. GLI1 Inhibitor SRI-38832 Attenuates Chemotherapeutic Resistance by Downregulating NBS1 Transcription in BRAFV600E Colorectal Cancer
- Author
-
Ruowen Zhang, Jinlu Ma, Justin T. Avery, Vijaya Sambandam, Theresa H. Nguyen, Bo Xu, Mark J. Suto, and Rebecca J. Boohaker
- Subjects
colorectal cancer ,5-FU resistance ,DNA damage repair ,GLI1 target gene ,novel GLI1 inhibitor ,target therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
4. HBV Core Promoter Inhibition by Tubulin Polymerization Inhibitor (SRI-32007)
- Author
-
Raj Kalkeri, Junzhong Peng, Chunsheng Huang, Zhaohui Cai, Roger G. Ptak, and Mark J. Suto
- Subjects
Microbiology ,QR1-502 - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
5. Supplementary Methods from Runx2 Deficiency in Osteoblasts Promotes Myeloma Resistance to Bortezomib by Increasing TSP-1–Dependent TGFβ1 Activation and Suppressing Immunity in Bone Marrow
- Author
-
Yang Yang, Juan Li, Joanne E. Murphy-Ullrich, Amjad Javed, Mark J. Suto, Yun Lu, Pramod S. Gowda, Timothy N. Trotter, Xiaoxuan Xu, and Chao Zhang
- Abstract
Supplementary MethodsSupplementary Methods descript detailed methods of Bioluminescence imaging, ELISA, Flow cytometry, Isolation and culture of Pre-OBs with 5TGM1-Luc MM cells, Generation and sorting of MDSCs, Cell proliferation assays, Treatment of MM cells with OB-Runx2+/+ and OB-Runx2-/- BMS in vitro, and Western blotting.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure S1-S6 from Runx2 Deficiency in Osteoblasts Promotes Myeloma Resistance to Bortezomib by Increasing TSP-1–Dependent TGFβ1 Activation and Suppressing Immunity in Bone Marrow
- Author
-
Yang Yang, Juan Li, Joanne E. Murphy-Ullrich, Amjad Javed, Mark J. Suto, Yun Lu, Pramod S. Gowda, Timothy N. Trotter, Xiaoxuan Xu, and Chao Zhang
- Abstract
Supplementary Figure S1-S6Supplementary Figure S1 shows tumor burden in the mice before BTZ treatment started (S1A), MM cell growth and apoptosis after co-cultured with BMS or pre-OBs of OB-Runx2-/- or OB-Runx2+/+ mice (S1B-F), and the proportion of PMN-MDSCs and M-MDSCs in the BM of Runx2-/- or OB-Runx2+/+ mice after 4-week treatment with BTZ (S1G-H).Supplementary Figure S2 shows tumor burden in the mice before SRI31277 and BTZ treatment (S2A) and the proportion of PMN-MDSCs and M-MDSCs in the BM of Runx2-/- or OB-Runx2+/+ mice after 4-week treatment with SRI31277 and BTZ (S2B-C).Supplementary Figure S3 shows tumor burden in the mice before 5-Fu and BTZ treatment.Supplementary Figure S4 shows the impact of GEM treatment on BTZ resistance of MM cells in OB-Runx2-/- mice.Supplementary Figure S5 shows the proportion of PMN-MDSCs and M-MDSCs in the BM of Runx2-/- or OB-Runx2+/+ mice after 4-week treatment with 5-Fu and BTZ.Supplementary Figure S6 shows the impact of GEM and BTZ treatment on MDSCs and CD8 T cells.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S1 from Runx2 Deficiency in Osteoblasts Promotes Myeloma Resistance to Bortezomib by Increasing TSP-1–Dependent TGFβ1 Activation and Suppressing Immunity in Bone Marrow
- Author
-
Yang Yang, Juan Li, Joanne E. Murphy-Ullrich, Amjad Javed, Mark J. Suto, Yun Lu, Pramod S. Gowda, Timothy N. Trotter, Xiaoxuan Xu, and Chao Zhang
- Abstract
Supplementary Table S1Supplementary Table S1 shows the company's name and the product numbers of Bortezomib, 5-Fluorouracil and Gemcitabine as well as the company's name and the product numbers of all antibodies listed in table S1, which we purchased and used for this study.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Runx2 Deficiency in Osteoblasts Promotes Myeloma Resistance to Bortezomib by Increasing TSP-1–Dependent TGFβ1 Activation and Suppressing Immunity in Bone Marrow
- Author
-
Yang Yang, Juan Li, Joanne E. Murphy-Ullrich, Amjad Javed, Mark J. Suto, Yun Lu, Pramod S. Gowda, Timothy N. Trotter, Xiaoxuan Xu, and Chao Zhang
- Abstract
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 TGFβ1 in the BM. In multiple myeloma tumor-bearing OB-Runx2−/− mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)–mediated TGFβ1 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.
- Published
- 2023
- Full Text
- View/download PDF
9. Runx2 Deficiency in Osteoblasts Promotes Myeloma Resistance to Bortezomib by Increasing TSP-1–Dependent TGFβ1 Activation and Suppressing Immunity in Bone Marrow
- Author
-
Chao Zhang, Xiaoxuan Xu, Timothy N. Trotter, Pramod S. Gowda, Yun Lu, Mark J. Suto, Amjad Javed, Joanne E. Murphy-Ullrich, Juan Li, and Yang Yang
- Subjects
Cancer Research ,Osteoblasts ,Core Binding Factor Alpha 1 Subunit ,Article ,Bortezomib ,Thrombospondin 1 ,Transforming Growth Factor beta1 ,Disease Models, Animal ,Mice ,Oncology ,Bone Marrow ,Cell Line, Tumor ,Animals ,Humans ,Multiple Myeloma - Abstract
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 TGFβ1 in the BM. In multiple myeloma tumor-bearing OB-Runx2−/− mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)–mediated TGFβ1 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.
- Published
- 2022
- Full Text
- View/download PDF
10. A small molecule that induces translational readthrough of CFTR nonsense mutations by eRF1 depletion
- Author
-
Ming Du, Ning Peng, Jamie R Wangen, Jianguo Chen, Rachel Green, Venkateshwar Mutyam, Yao Li, Kari Thrasher, Corinne E. Augelli-Szafran, Jerome Mahiou, Feng Liang, Jyoti Sharma, Lianwu Fu, Candice J. Coppola, Kim M. Keeling, Martin Mense, Eric M. Mendenhall, Jeong S. Hong, Hermann Bihler, Liping Tang, Robert Bostwick, Eric J. Sorscher, Bini Mathew, Josef Saltz, Steven M. Rowe, Andras Rab, Kaimao Liu, David M. Bedwell, Mark J. Suto, and Eric Wong
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Termination factor ,viruses ,Science ,Nonsense mutation ,Nonsense-mediated decay ,General Physics and Astronomy ,Cystic Fibrosis Transmembrane Conductance Regulator ,RNA decay ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cystic fibrosis ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,RNA interference ,Genes, Reporter ,Humans ,Multidisciplinary ,Molecular medicine ,Chemistry ,Translational readthrough ,HEK 293 cells ,fungi ,Translation (biology) ,Epithelial Cells ,General Chemistry ,Peptide Chain Termination, Translational ,Stop codon ,Cell biology ,Nonsense Mediated mRNA Decay ,030104 developmental biology ,Aminoglycosides ,HEK293 Cells ,Codon, Nonsense ,Microsomes, Liver ,RNA ,RNA Interference ,Gentamicins ,Ribosomes ,030217 neurology & neurosurgery ,Peptide Termination Factors - Abstract
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., Premature termination codons can cause early translation termination and lead to disease. Here the authors perform a screen to identify compounds with readthrough activity and show that these reduce eRF1 levels to suppress premature termination associated with cystic fibrosis.
- Published
- 2021
11. Targeting Chikungunya Virus Replication by Benzoannulene Inhibitors
- Author
-
Mark T. Heise, Thomas E. Morrison, Subramaniam Ananthan, Joseph A. Maddry, Sanjay Sarkar, Jessica L. Smith, Kevin J Rodzinak, Nicole N. Haese, Daniel N. Streblow, Nathaniel J. Moorman, Cassilyn E. Streblow, Ashish K. Pathak, Vibha Pathak, Aaron D Streblow, Lynn Rasmussen, Fahim Ahmad, Corinne E. Augelli-Szafran, S Kaleem Ahmed, Victor R. DeFilippis, Robert Bostwick, Craig N. Kreklywich, Mark J. Suto, Sixue Zhang, Robbie Allen, Alec J. Hirsch, Jaden T Cowan, Babu L. Tekwani, Kiley Bonin, Nichole A. Tower, Wes Sander, Valerie J Smith, Mousheng Wu, John M Gerdes, Clayton R. Morrison, and Omar Moukha-Chafiq
- Subjects
Male ,Oxidoreductases Acting on CH-CH Group Donors ,Cell Survival ,Dihydroorotate Dehydrogenase ,Virus Replication ,Antiviral Agents ,01 natural sciences ,Article ,Virus ,Cell Line ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Drug Discovery ,Benzene Derivatives ,medicine ,Animals ,Humans ,Benzamide ,Cytotoxicity ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Chemistry ,virus diseases ,Virology ,0104 chemical sciences ,Mice, Inbred C57BL ,Molecular Docking Simulation ,Disease Models, Animal ,010404 medicinal & biomolecular chemistry ,Titer ,Mechanism of action ,Viral replication ,Microsomes, Liver ,Dihydroorotate dehydrogenase ,Chikungunya Fever ,Molecular Medicine ,Female ,medicine.symptom ,Chikungunya virus ,Half-Life - Abstract
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 EC(90) = 1.45 μM and viral titer reduction (VTR) of 2.5 log at 10 μM with no observed cytotoxicity (CC(50) = 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 (EC(90) = 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.
- Published
- 2021
- Full Text
- View/download PDF
12. HBV Core Promoter Inhibition by Tubulin Polymerization Inhibitor (SRI-32007)
- Author
-
Roger G. Ptak, Mark J. Suto, Zhaohui Cai, Chunsheng Huang, Raj Kalkeri, and Junzhong Peng
- Subjects
0301 basic medicine ,Article Subject ,Cell ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,In vivo ,Virology ,Genotype ,medicine ,Hepatitis B virus ,virus diseases ,Promoter ,medicine.disease ,QR1-502 ,digestive system diseases ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Research Article - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
13. Development of small‐molecule tau‐SH3 interaction inhibitors that prevent amyloid‐β toxicity
- Author
-
Jonathan R Roth, Travis Rush, Derian A Pugh, Hunter B Dean, J. Nicholas Cochran, Samantha J Thompson, Adam R Aldaher, Jacob S Mesina, Pedro Ruiz, Vibha Pathak, Mousheng Wu, J Robert Bostwick, Mark J Suto, Corinne E Augelli‐Szafran, and Erik D. Roberson
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
14. Identification of Quinolinones as Antivirals against Venezuelan Equine Encephalitis Virus
- Author
-
Nathaniel J. Moorman, Nikhil Madadi, Daniel N. Streblow, Nicole N. Haese, Robert Bostwick, Mark T. Heise, Wes Sanders, Theresa H Nguyen, Corinne E. Augelli-Szafran, Nichole A Towers, Omar Moukha-Chafiq, Victor R. DeFilippis, Joseph A. Maddry, Sixue Zhang, Ashish K. Pathak, Mark J. Suto, Subramaniam Ananthan, Sharon Taft-Benz, Lynn Rasmussen, Shuklendu D Karyakarte, Michael Denton, Nicholas A May, Kevin J Rodzinak, Aaron D Streblow, and Thomas E. Morrison
- Subjects
viruses ,Alphavirus ,Quinolones ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Virus ,Encephalitis Virus, Venezuelan Equine ,03 medical and health sciences ,Ross River virus ,0302 clinical medicine ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Chikungunya ,Horses ,Alphavirus infection ,030304 developmental biology ,030203 arthritis & rheumatology ,Pharmacology ,0303 health sciences ,biology ,virus diseases ,biology.organism_classification ,medicine.disease ,Virology ,Human morbidity ,Infectious Diseases ,Viral replication ,Venezuelan equine encephalitis virus ,Chikungunya virus - Abstract
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.
- Published
- 2021
15. Inhibition of the SET/MLL Histone Methyltransferase Complex as a Novel Epigenetic Targeted Therapy in Mixed Lineage Leukemia
- Author
-
Robert Bostwick, Corinne Augello-Szafran, William J. Placzek, Hayley Widden, Anish Vadukoot, Rebecca J. Boohaker, Mark J. Suto, Omar Moukha-Chafiq, and Robert H. Whitaker
- Subjects
MIXED LINEAGE LEUKEMIA ,medicine.medical_treatment ,Genetics ,Cancer research ,medicine ,Histone methyltransferase complex ,Epigenetics ,Biology ,Molecular Biology ,Biochemistry ,Biotechnology ,Targeted therapy - Published
- 2021
- Full Text
- View/download PDF
16. Development of BRD4 inhibitors as anti-inflammatory agents and antidotes for arsenicals
- Author
-
Marina Fosso, Yatchang, Bini, Mathew, Ritesh K, Srivastava, Jasim, Khan, Suhail, Muzaffar, Sixue, Zhang, Mousheng, Wu, Ling, Zhai, Pedro, Ruiz, Anupam, Agarwal, James R, Bostwick, Mark J, Suto, Mohammad, Athar, and Corinne E, Augelli-Szafran
- Subjects
Antidotes ,Organic Chemistry ,Clinical Biochemistry ,Drug Discovery ,Anti-Inflammatory Agents ,Nuclear Proteins ,Pharmaceutical Science ,Molecular Medicine ,Molecular Biology ,Biochemistry ,Arsenicals ,Article ,Transcription Factors - Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
17. New drug hope for diabetes mellitus
- Author
-
Peng Li, Junqin Chen, Anath Shalev, Jason K. Kim, Guanlan Xu, Lance A. Thielen, Omar Moukha-Chafiq, Praveen Sethupathy, Seong Ho Jo, Truman Grayson, Mark J. Suto, Gu Jing, Corinne E. Augelli-Szafran, Brian Lu, and Matt Kanke
- Subjects
Male ,0301 basic medicine ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Administration, Oral ,Pharmacology ,Alpha cell ,Mice ,Endocrinology ,0302 clinical medicine ,Cells, Cultured ,media_common ,geography.geographical_feature_category ,Chemistry ,Glucagon secretion ,Islet ,Beta cell ,TXNIP ,medicine.drug ,Drug ,medicine.medical_specialty ,media_common.quotation_subject ,MEDLINE ,Glucagon ,Streptozocin ,Article ,Diabetes Mellitus, Experimental ,Small Molecule Libraries ,03 medical and health sciences ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Hypoglycemic Agents ,Humans ,Molecular Biology ,geography ,business.industry ,Insulin ,Cell Biology ,medicine.disease ,Streptozotocin ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Carrier protein ,business ,Carrier Proteins ,030217 neurology & neurosurgery - Abstract
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 mice) 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.
- Published
- 2020
18. Runx2 deficiency in osteoblasts promotes myeloma resistance to bortezomib by increasing thrombospondin 1-mediated TGF-ß1 activation and suppression immunity in the bone marrow
- Author
-
Chao Zhang, Xiaoxuan Xu, Timothy N. Trotter, Pramod S. Gowda, Yun Lu, Mark J. Suto, Amjad Javed, Joanne E. Murphy-Ullrich, Juan Li, and Yang Yang
- Abstract
Background: Multiple myeloma (MM) is a plasma cell malignancy that thrives in the bone marrow (BM). Although the proteasome inhibitor bortezomib (BTZ) is one of the most effective front-line chemotherapeutic drugs for MM, 15–20% of high-risk patients do not respond to this drug or become resistant to treatment. The mechanisms driving this chemoresistance remain unclear. Previous studies showed that the tumor microenvironment contributes to cancer chemoresistance. Our recent studies demonstrated that Runt-related transcription factor 2 (Runx2) deficiency in osteoblasts (OBs) creates a cytokine-rich and immunosuppressive microenvironment in the BM and promotes MM progression. However, the impact of Runx2 deficiency in OBs on the efficacy of BTZ in treatment of MM is still unknown. Methods: We assessed the effects of OB-Runx2 deficiency on the outcome of BTZ treatment in OB-Runx2 +/+ and OB-Runx2 -/- mouse models with MM using bioluminescence imaging, ELISA and flow cytometry. In addition, we used a co-culturing in vitro system to explore the mechanism of BTZ resistance and assessed this system by MTT assays and Western blot analysis. Results: We discovered that OB-Runx2 deficiency induces MM cells resistance to BTZ via the suppression of immunity and increased active TGF-β1 in the BM. We further demonstrated that depletion of myeloid-derived suppressor cells (MDSCs) by gemcitabine or inhibition of TGF-ß1 activity by SRI31277, a compound that blocks thrombospondin 1 (TSP1)-mediated TGF-ß1 activation, restores anti-tumor immunity in the BM and overcomes BTZ resistance induced by OB-Runx2 deficiency. In addition, SRI31277 also directly inhibits the activity of canonical (Smad2/3) and non-canonical (Erk1/2) signaling pathways of TGF-ß1 in 5TGM1-Luc MM cells and sensitizes MM cells to BTZ, resulting in increased apoptosis of MM cells. Conclusions: OB-Runx2 deficiency promotes BTZ resistance in MM cells through the regulations of MDSCs and TSP1-mediated TGF-ß1 activation in the BM. These data identify novel mechanisms of BTZ resistance in MM and suggest new strategies to overcome BTZ resistance in treatment of MM.
- Published
- 2020
- Full Text
- View/download PDF
19. Identification of Inhibitors of Thrombospondin 1 Activation of TGF-β
- Author
-
Vandana V. Gupta, Manuel A. Pallero, Joanne E. Murphy-Ullrich, Wei Zhang, Mark J. Suto, and Bini Mathew
- Subjects
010405 organic chemistry ,Chemistry ,Organic Chemistry ,Matricellular protein ,Context (language use) ,medicine.disease ,Ligand (biochemistry) ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,In vivo ,Fibrosis ,Drug Discovery ,Thrombospondin 1 ,Cancer research ,medicine ,Kinase activity ,Receptor - Abstract
[Image: see text] TGF-β has been a target of interest for the treatment of fibrotic diseases and certain cancers. Approaches to target TGF-β include antagonists of the active ligand or TGF-β 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-β are needed. We previously reported that the matricellular protein, thrombospondin 1, activates the latent TGF-β 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-β activation (pIC(50) = 8.12 nM) and has a plasma half life of 1.8 h (iv).
- Published
- 2020
20. GLI1 Inhibitor SRI-38832 Attenuates Chemotherapeutic Resistance by Downregulating NBS1 Transcription in BRAFV600E Colorectal Cancer
- Author
-
Theresa H Nguyen, Rebecca J. Boohaker, Ruowen Zhang, Mark J. Suto, Bo Xu, Vijaya Sambandam, Justin T. Avery, and Jinlu Ma
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,DNA damage ,medicine.medical_treatment ,Regulator ,colorectal cancer ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,GLI1 ,In vivo ,medicine ,DNA damage repair ,Original Research ,Chemotherapy ,integumentary system ,biology ,target therapy ,business.industry ,novel GLI1 inhibitor ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,GLI1 target gene ,In vitro ,3. Good health ,030104 developmental biology ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business ,5-FU resistance - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
21. Development and application of high-throughput screens for the discovery of compounds that disrupt ErbB4 signaling: Candidate cancer therapeutics
- Author
-
Laura J. Cook, Ram B. Gupta, Christopher P. Mill, Richard L. Cullum, Cristina C. Rael, Megan A. Jacobi, Logan Neel, Ling Zhai, Mark J. Suto, Allan E. David, Robert Bostwick, Jared I. Senfeld, Lauren M. Lucas, Forrest Smith, Kanupriya Whig, David J. Riese, Darby C. Taylor, Mackenzie H. Harris, John T. Piazza, and David P. Kaufmann
- Subjects
0301 basic medicine ,Partial Agonists ,Receptor, ErbB-4 ,Biochemistry ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Aromatic Amino Acids ,0302 clinical medicine ,Drug Discovery ,Medicine and Health Sciences ,Enzyme assays ,Colorimetric assays ,Post-Translational Modification ,Phosphorylation ,Amino Acids ,Enzyme-Linked Immunoassays ,Bioassays and physiological analysis ,Melanoma ,ERBB4 ,MTT assay ,Multidisciplinary ,Organic Compounds ,Drug discovery ,Drugs ,Chemistry ,Optical Equipment ,Cell Processes ,Gain of Function Mutation ,030220 oncology & carcinogenesis ,Physical Sciences ,Engineering and Technology ,Medicine ,Research Article ,Biotechnology ,Signal Transduction ,Agonist ,medicine.drug_class ,Science ,Equipment ,Bioengineering ,Biology ,Partial agonist ,Small Molecule Libraries ,03 medical and health sciences ,Hydroxyl Amino Acids ,Cell Line, Tumor ,medicine ,Humans ,Nerve Growth Factors ,Immunoassays ,Cell Proliferation ,Pharmacology ,Cell growth ,Organic Chemistry ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,Prisms ,Tyrosine phosphorylation ,Cell Biology ,Research and analysis methods ,030104 developmental biology ,chemistry ,Small Molecules ,Biochemical analysis ,Immunologic Techniques ,Cancer research ,biology.protein ,Tyrosine - Abstract
Whereas recent clinical studies report metastatic melanoma survival rates high as 30–50%, many tumors remain nonresponsive or become resistant to current therapeutic strategies. Analyses of The Cancer Genome Atlas (TCGA) skin cutaneous melanoma (SKCM) data set suggests that a significant fraction of melanomas potentially harbor gain-of-function mutations in the gene that encodes for the ErbB4 receptor tyrosine kinase. In this work, a drug discovery strategy was developed that is based on the observation that the Q43L mutant of the naturally occurring ErbB4 agonist Neuregulin-2beta (NRG2β) functions as a partial agonist at ErbB4. NRG2β/Q43L stimulates tyrosine phosphorylation, fails to stimulate ErbB4-dependent cell proliferation, and inhibits agonist-induced ErbB4-dependent cell proliferation. Compounds that exhibit these characteristics likely function as ErbB4 partial agonists, and as such hold promise as therapies for ErbB4-dependent melanomas. Consequently, three highly sensitive and reproducible (Z’ > 0.5) screening assays were developed and deployed for the identification of small-molecule ErbB4 partial agonists. Six compounds were identified that stimulate ErbB4 phosphorylation, fail to stimulate ErbB4-dependent cell proliferation, and appear to selectively inhibit ErbB4-dependent cell proliferation. Whereas further characterization is needed to evaluate the full therapeutic potential of these molecules, this drug discovery platform establishes reliable and scalable approaches for the discovery of ErbB4 inhibitors.
- Published
- 2020
22. Identification of Cosalane as an Inhibitor of Human and Murine CC–Chemokine Receptor 7 Signaling via a High-Throughput Screen
- Author
-
James M. Coghill, Dmitri Kireev, Mark J. Suto, Emily A. Hull-Ryde, Kenneth H. Pearce, Kenneth A. Fowler, Kelin Li, Melissa A. Porter, Maria F. Sassano, William P. Janzen, and Catherine Simpson
- Subjects
0301 basic medicine ,Receptors, CCR7 ,T-Lymphocytes ,chemical and pharmacologic phenomena ,C-C chemokine receptor type 7 ,Ligands ,01 natural sciences ,Biochemistry ,Cell Line ,Receptors, G-Protein-Coupled ,Analytical Chemistry ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Immune system ,Animals ,Humans ,Receptor ,G protein-coupled receptor ,Molecular Structure ,Chemistry ,Chemotaxis ,Aurintricarboxylic Acid ,CCL19 ,hemic and immune systems ,High-Throughput Screening Assays ,0104 chemical sciences ,Cell biology ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,Drug Design ,Molecular Medicine ,CC chemokine receptors ,Signal Transduction ,Biotechnology ,CCL21 - Abstract
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 β-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.
- Published
- 2018
- Full Text
- View/download PDF
23. Structure–activity relationships and cellular mechanism of action of small molecules that enhance the delivery of oligonucleotides
- Author
-
Mark J. Suto, Xin Ming, Edward G Brown, Lindsey I. James, Rudolph L. Juliano, Chengqiong Mao, Ling Wang, Yamuna Ariyarathna, and Francis X. Tavares
- Subjects
0301 basic medicine ,Pyridines ,Oligonucleotide ,Endosome ,Oligonucleotides ,Intracellular Membranes ,Biology ,Small molecule ,Cell biology ,Cell membrane ,Structure-Activity Relationship ,03 medical and health sciences ,Cytosol ,030104 developmental biology ,medicine.anatomical_structure ,Chemical Biology and Nucleic Acid Chemistry ,Pyrazines ,Genetics ,medicine ,Humans ,Structure–activity relationship ,Endomembrane system ,Intracellular ,HeLa Cells - Abstract
The pharmacological effects of antisense and siRNA oligonucleotides are hindered by the tendency of these molecules to become entrapped in endomembrane compartments thus failing to reach their targets in the cytosol or nucleus. We have previously used high throughput screening to identify small molecules that enhance the escape of oligonucleotides from intracellular membrane compartments and have termed such molecules OECs (oligonucleotide enhancing compounds). Here, we report on the structure–activity relationships of a family of OECs that are analogs of a hit that emerged from our original screen. These studies demonstrate key roles for the lipophilic aromatic groups, the tertiary nitrogen, and the carbamate moiety of the parent compound. We have also investigated the intracellular site of action of the OECs and have shown that activity is due to the release of oligonucleotides from intermediate endosomal compartments rather than from early endosomes or from highly acidic downstream compartments. At high concentrations of OECs toxicity occurs in a manner that is independent of caspases or of lysosomal cathepsins but instead involves increased plasma membrane permeability. Thus, in addition to describing specific characteristics of this family of OECs, the current study provides insights into basic mechanisms of oligonucleotide trafficking and their implications for oligonucleotide delivery.
- Published
- 2018
- Full Text
- View/download PDF
24. Predicting radiotherapy response for patients with soft tissue sarcoma by developing a molecular signature
- Author
-
Xinyan Zhang, Mark J. Suto, Nengjun Yi, Bo Xu, Yan Li, Zaixiang Tang, and Qinghua Zeng
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,sarcoma ,medicine.medical_treatment ,Biology ,gene signature ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Databases, Genetic ,medicine ,Cluster Analysis ,Humans ,Gene Regulatory Networks ,Radiosensitivity ,Survival analysis ,Aged ,Sequence Analysis, RNA ,Soft tissue sarcoma ,Computational Biology ,treatment response ,Soft tissue ,Cancer ,Articles ,prediction ,General Medicine ,Middle Aged ,Gene signature ,medicine.disease ,Survival Analysis ,Molecular medicine ,3. Good health ,Gene Expression Regulation, Neoplastic ,Radiation therapy ,Treatment Outcome ,030104 developmental biology ,radiosensitivity ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Female ,Radiotherapy, Adjuvant - Abstract
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.
- Published
- 2017
- Full Text
- View/download PDF
25. Discovery of novel frizzled-7 inhibitors by targeting the receptor’s transmembrane domain
- Author
-
Subramaniam Ananthan, Yonghe Li, Mark J. Suto, Wei Zhang, and Wenyan Lu
- Subjects
0301 basic medicine ,Wnt/β-catenin ,Virtual screening ,Frizzled ,business.industry ,Drug discovery ,Wnt signaling pathway ,Cancer ,virtual screening ,Bioinformatics ,medicine.disease ,3. Good health ,inhibitor ,03 medical and health sciences ,Transmembrane domain ,030104 developmental biology ,Oncology ,Cancer research ,cancer ,Medicine ,frizzled ,business ,Receptor ,Research Paper ,G protein-coupled receptor - Abstract
// Wei Zhang 1 , Wenyan Lu 2 , Subramaniam Ananthan 1 , Mark J. Suto 1 and Yonghe Li 2 1 Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States 2 Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, Alabama 35205, United States Correspondence to: Wei Zhang, email: zhangwee@yahoo.com Yonghe Li, email: yli@southernresearch.org Keywords: frizzled, Wnt/β-catenin, inhibitor, virtual screening, cancer Received: March 29, 2017 Accepted: June 28, 2017 Published: September 06, 2017 ABSTRACT 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 β-catenin pathway which is important for both initiation and progression of cancers. Disruption of the Wnt/β-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 IC 50 values in the sub-micromolar range and inhibited cancer cell proliferation with IC 50 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.
- Published
- 2017
- Full Text
- View/download PDF
26. Repositioning chlorpromazine for treating chemoresistant glioma through the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory subunit
- Author
-
Corinne E. Griguer, Mark J. Suto, Claudia R. Oliva, Wei Zhang, and Cathy Langford
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Chlorpromazine ,Brain tumor ,Mice, Nude ,Pharmacology ,Mitochondria, Heart ,Electron Transport Complex IV ,03 medical and health sciences ,stem cells ,Glioma ,cytochrome c oxidase ,Cell Line, Tumor ,medicine ,Temozolomide ,Animals ,Humans ,Antineoplastic Agents, Alkylating ,Cell Proliferation ,Cell growth ,business.industry ,Brain Neoplasms ,glioblastoma ,Drug Repositioning ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,inhibitor ,Dacarbazine ,030104 developmental biology ,Oncology ,Cell culture ,Drug Resistance, Neoplasm ,Dopamine Antagonists ,Cattle ,Stem cell ,business ,medicine.drug ,Research Paper ,Antipsychotic Agents - Abstract
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.
- Published
- 2017
27. Development of a radiosensitivity gene signature for patients with soft tissue sarcoma
- Author
-
Zaixiang Tang, Xinyan Zhang, Mark J. Suto, Qinghua Zeng, Bo Xu, Nengjun Yi, Yan Li, and Jinlu Ma
- Subjects
Male ,0301 basic medicine ,Oncology ,Pathology ,sarcoma ,Databases, Factual ,medicine.medical_treatment ,gene signature ,Radiation Tolerance ,0302 clinical medicine ,Histologic type ,Cluster Analysis ,survival prediction ,Aged, 80 and over ,Soft tissue sarcoma ,Middle Aged ,Prognosis ,Combined Modality Therapy ,Tumor Burden ,3. Good health ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Sarcoma ,Research Paper ,Adult ,medicine.medical_specialty ,radio-sensitivity ,Young Adult ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,radiotherapy ,Aged ,business.industry ,Gene Expression Profiling ,Disease progression ,Computational Biology ,Reproducibility of Results ,Gene signature ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Genetic epidemiology ,Radiotherapy, Adjuvant ,Biostatistics ,Transcriptome ,business - Abstract
// Zaixiang Tang 1, 2, 3, 4 , Qinghua Zeng 5 , Yan Li 4 , Xinyan Zhang 4 , Jinlu Ma 5, 6 , Mark J. Suto 5 , Bo Xu 5 , Nengjun Yi 4 1 Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou 215123, China 2 Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, 215123, China 3 Center for Genetic Epidemiology and Genomics, Medical College of Soochow University, Suzhou, 215123, China 4 Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA 5 Drug Discovery Division, Southern Research Institute, Birmingham, AL 35294, USA 6 Department of Radiation Oncology, The First Hospital, Xi’an Jiaotong University, Xi’an, Shanxi, 710061, China Correspondence to: Nengjun Yi, email: nyi@uab.edu Bo Xu, email: bxu@southernresearch.org , bxu@uab.edu Keywords: gene signature, radio-sensitivity, radiotherapy, survival prediction, sarcoma Received: September 27, 2016 Accepted: January 24, 2017 Published: March 15, 2017 ABSTRACT Adjuvant radiotherapy is an important clinical treatment option for the majority of sarcomas. The motivation of current study is to identify a gene signature and to predict radiosensitive patients who are most likely to benefit from radiotherapy. Using the public available data of soft tissue sarcoma from The Cancer Genome Atlas, we developed a cross-validation procedure for identifying a gene signature and predicting radiosensitive patients through. The result showed that the predicted radiosensitive patients who received radiotherapy had a significantly better survival with a reduced rate of new tumor event and disease progression. Strata analysis showed that the predicted radiosensitive patients had significantly better survival under radiotherapy independent of histologic types. A hierarchical cluster analysis was used to validate the gene signature, and the results showed the predicted sensitivity for each patient well matched the results from cluster analysis. Together, we demonstrate a radiosensitive molecular signature that can be potentially used for identifying radiosensitive patients with sarcoma.
- Published
- 2017
- Full Text
- View/download PDF
28. RETRACTED: SRI36160 is a specific inhibitor of Wnt/β-catenin signaling in human pancreatic and colorectal cancer cells
- Author
-
Patsy G. Oliver, Mark J. Suto, Wenyan Lu, Sivaram Sridharan, Corinne E. Augelli-Szafran, Donald J. Buchsbaum, Vibha Pathak, and Yonghe Li
- Subjects
0301 basic medicine ,Cancer Research ,Frizzled ,medicine.medical_specialty ,Adenomatous polyposis coli ,Colorectal cancer ,medicine.medical_treatment ,Adenomatous Polyposis Coli Protein ,Article ,Targeted therapy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Wnt3A Protein ,Internal medicine ,Pancreatic cancer ,medicine ,Animals ,Humans ,Phosphorylation ,STAT3 ,Wnt Signaling Pathway ,Cells, Cultured ,beta Catenin ,Cell Proliferation ,biology ,Wnt signaling pathway ,LRP6 ,medicine.disease ,Pancreatic Neoplasms ,Wnt Proteins ,030104 developmental biology ,Endocrinology ,Oncology ,Low Density Lipoprotein Receptor-Related Protein-6 ,030220 oncology & carcinogenesis ,Mutation ,biology.protein ,Cancer research ,Benzimidazoles ,Colorectal Neoplasms - Abstract
Activation of Wnt/β-catenin signaling is associated with pancreatic and colorectal cancer, among others. To-date, there are no FDA-approved small molecule Wnt/β-catenin inhibitors and many past efforts resulted in compounds with undesirable off-target effects. We recently identified a series of benzimidazole analogs as potent inhibitors of Wnt/β-catenin signaling. Here, we show that the lead compound SRI36160 displayed selective Wnt inhibition and potent antiproliferative activity in pancreatic and colorectal cancer cells. Moreover, SRI36160 had no effect on STAT3 and mTORC1 signaling in pancreatic and colorectal cancer cells, and was not effective in inhibiting proliferation of non-cancerous cells. Our findings suggest that this series of benzimidazole analogs presents a novel approach for the treatment of Wnt-dependent cancers such as colorectal and pancreatic cancer.
- Published
- 2017
- Full Text
- View/download PDF
29. Development of novel small molecules for the treatment of ALS
- Author
-
Pedro Ruiz, Vibha Pathak, Bini Mathew, and Mark J. Suto
- Subjects
Clinical Biochemistry ,SOD1 ,SOD2 ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Superoxide dismutase ,Small Molecule Libraries ,chemistry.chemical_compound ,Structure-Activity Relationship ,Superoxide Dismutase-1 ,In vivo ,Drug Discovery ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Molecular Biology ,Gene ,biology ,010405 organic chemistry ,Chemistry ,Superoxide Dismutase ,Organic Chemistry ,Amyotrophic Lateral Sclerosis ,NF-kappa B ,NF-κB ,medicine.disease ,In vitro ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Gene Expression Regulation ,biology.protein ,Cancer research ,Molecular Medicine - Abstract
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.
- Published
- 2019
30. Triamterene normalizes glycosaminoglycan accumulation in an IDUA-W402X mouse model of MPS I (Hurler syndrome) via nonsense suppression
- Author
-
Josh Echols, Ming Du, David M. Bedwell, Lynn Rasmussen, Mark J. Suto, Amna Siddiqui, J. Robert Bostwick, Kim M. Keeling, and Halil Dündar
- Subjects
Triamterene ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,media_common.quotation_subject ,Nonsense ,medicine.disease ,Biochemistry ,Glycosaminoglycan ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Hurler syndrome ,business ,Molecular Biology ,medicine.drug ,media_common - Published
- 2019
31. Studies on Dibenzylamines as Inhibitors of Venezuelan Equine Encephalitis Virus
- Author
-
Victor R. DeFilippis, Theresa H Nguyen, Mark J. Suto, Corinne E. Augelli-Szafran, Sharon Taft-Benz, Kiley Bonin, Lynn Rasmussen, Cassilyn E. Streblow, Daniel N. Streblow, Sanjay Sarkar, Ashish K. Pathak, Nichole A. Tower, Nicole N. Haese, Thomas E. Morrison, Nikhil Madadi, Mark T. Heise, and Robert Bostwick
- Subjects
0301 basic medicine ,Benzylamines ,viruses ,030106 microbiology ,Cell ,Disease ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Virus ,Cell Line ,Encephalitis Virus, Venezuelan Equine ,Small Molecule Libraries ,03 medical and health sciences ,Chlorocebus aethiops ,medicine ,Protein biosynthesis ,Animals ,Humans ,Vero Cells ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,business.industry ,Encephalomyelitis, Venezuelan Equine ,Viral Load ,biology.organism_classification ,Phenotype ,Virology ,High-Throughput Screening Assays ,Titer ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Venezuelan equine encephalitis virus ,Togaviridae ,business - Abstract
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.
- Published
- 2019
- Full Text
- View/download PDF
32. Structure-activity relationship (SAR) studies of N-(3-methylpyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (SRI-22819) as NF-ҡB activators for the treatment of ALS
- Author
-
Sixue Zhang, Pedro Ruiz, Rita M. Cowell, Kaval D. Patel, Shilpa Dutta, Mark J. Suto, Bini Mathew, Micah S. Simmons, Corinne E. Augelli-Szafran, and Jordan T. Entrekin
- Subjects
Duchenne muscular dystrophy ,SOD1 ,SOD2 ,Context (language use) ,Pharmacology ,Cell Line ,Polar surface area ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,ADME ,Oxadiazoles ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Organic Chemistry ,NF-kappa B ,General Medicine ,medicine.disease ,Ataluren ,Molecular Docking Simulation ,chemistry - Abstract
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-ҡBs. We previously reported that SRI-22819 increases NF-ҡB 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.
- Published
- 2021
- Full Text
- View/download PDF
33. Consortia's critical role in developing medical countermeasures for re-emerging viral infections: a USA perspective
- Author
-
Mark J. Suto, Richard J. Whitley, George R. Painter, and Maaike Everts
- Subjects
0301 basic medicine ,Government ,Economic growth ,Public fund ,business.industry ,West Nile virus ,Perspective (graphical) ,medicine.disease_cause ,medicine.disease ,Article ,03 medical and health sciences ,Reward system ,030104 developmental biology ,Virology ,Medicine ,Academic community ,Middle East respiratory syndrome ,business ,Pharmaceutical industry - Abstract
Viral infections, such as Ebola, severe acute respiratory syndrome/Middle East respiratory syndrome and West Nile virus have emerged as a serious health threat with no effective therapies. These infections have little commercial potential and are not a high priority for the pharmaceutical industry. However, the academic community has been active in this area for many years. The challenge is how to take this academic virology knowledge into a drug discovery and development domain. One approach is the use of consortia and public–private partnerships – this article highlights ongoing efforts in the USA. Public funds, such as those from government sources, can support research efforts that do not to appear to have commercial value. The key to success is finding a way to combine the different cultural and operational values and reward systems into a productive collaboration to identify new antivirals.
- Published
- 2016
- Full Text
- View/download PDF
34. Preferential Inhibition of Wnt/β-Catenin Signaling by Novel Benzimidazole Compounds in Triple-Negative Breast Cancer
- Author
-
Vibha Pathak, Mark J. Suto, Donald J. Buchsbaum, Patsy G. Oliver, Abhishek Gangrade, Han-Xun Wei, and Corinne E. Augelli-Szafran
- Subjects
0301 basic medicine ,LRP6 ,STAT3 Transcription Factor ,receptor-targeted therapy ,triple-negative breast cancer ,Wnt/β-catenin signaling ,benzimidazoles ,Notch signaling pathway ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Cyclin D1 ,Cell Line, Tumor ,Survivin ,Cytotoxic T cell ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Wnt Signaling Pathway ,Spectroscopy ,PI3K/AKT/mTOR pathway ,Triple-negative breast cancer ,beta Catenin ,Receptors, Notch ,Chemistry ,TOR Serine-Threonine Kinases ,Organic Chemistry ,Wnt signaling pathway ,General Medicine ,3. Good health ,Computer Science Applications ,Wnt Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Low Density Lipoprotein Receptor-Related Protein-6 ,Cancer research - Abstract
Wnt/β-catenin signaling is upregulated in triple-negative breast cancer (TNBC) compared to other breast cancer subtypes and normal tissues. Current Wnt/β-catenin inhibitors, such as niclosamide, target the pathway nonspecifically and exhibit poor pharmacokinetics/pharmacodynamics in vivo. Niclosamide targets other pathways, including mTOR, STAT3 and Notch. Novel benzimidazoles have been developed to inhibit Wnt/β-catenin signaling with greater specificity. The compounds SRI33576 and SRI35889 were discovered to produce more cytotoxicity in TNBC cell lines than in noncancerous cells. The agents also downregulated Wnt/β-catenin signaling mediators LRP6, cyclin D1, survivin and nuclear active β-catenin. In addition, SRI33576 did not affect mTOR, STAT3 and Notch signaling in TNBC and noncancerous cells. SRI35889 inhibited mTOR signaling less in noncancerous than in cancerous cells, while not affecting STAT3 and Notch pathways. Compounds SRI32529, SRI35357 and SRI35361 were not selectively cytotoxic against TNBC cell lines compared to MCF10A cells. While SRI32529 inhibited Wnt/β-catenin signaling, the compound also mitigated mTOR, STAT3 and Notch signaling. SRI33576 and SRI35889 were identified as cytotoxic and selective inhibitors of Wnt/β-catenin signaling with therapeutic potential to treat TNBC in vivo.
- Published
- 2018
35. Retraction notice to 'SRI36160 is a specific inhibitor of Wnt/beta-catenin signaling in human pancreatic and colorectal cancer cells' [Canc. Lett. 389C (2017) 41-48]
- Author
-
Mark J. Suto, Patsy G. Oliver, Yonghe Li, Corinne E. Augelli-Szafran, Donald J. Buchsbaum, Sivaram Sridharan, Wenyan Lu, and Vibha Pathak
- Subjects
Cancer Research ,Text mining ,Oncology ,Notice ,Colorectal cancer ,business.industry ,Catenin ,medicine ,Wnt signaling pathway ,Cancer research ,medicine.disease ,business ,Article - Abstract
Activation of Wnt/β-catenin signaling is associated with pancreatic and colorectal cancer, among others. To-date, there are no FDA-approved small molecule Wnt/β-catenin inhibitors and many past efforts resulted in compounds with undesirable off-target effects. We recently identified a series of benzimidazole analogs as potent inhibitors of Wnt/β-catenin signaling. Here, we show that the lead compound SRI36160 displayed selective Wnt inhibition and potent antiproliferative activity in pancreatic and colorectal cancer cells. Moreover, SRI36160 had no effect on STAT3 and mTORC1 signaling in pancreatic and colorectal cancer cells, and was not effective in inhibiting proliferation of non-cancerous cells. Our findings suggest that this series of benzimidazole analogs presents a novel approach for the treatment of Wnt-dependent cancers such as colorectal and pancreatic cancer.
- Published
- 2018
36. Rational design and development of a peptide inhibitor for the PD-1/PD-L1 interaction
- Author
-
Isaac Segura, Vijaya Sambandam, Mark J. Suto, James Miller, Bo Xu, and Rebecca J. Boohaker
- Subjects
0301 basic medicine ,Cancer Research ,Programmed Cell Death 1 Receptor ,Peptide ,Apoptosis ,Jurkat cells ,B7-H1 Antigen ,03 medical and health sciences ,Jurkat Cells ,Mice ,Cell Line, Tumor ,Blocking antibody ,Cytotoxic T cell ,Animals ,Humans ,Amino Acid Sequence ,Lymphocytes ,Cells, Cultured ,chemistry.chemical_classification ,biology ,Chemistry ,Rational design ,Cell biology ,Molecular Docking Simulation ,CTL ,030104 developmental biology ,Oncology ,Drug Design ,biology.protein ,Apoptotic signaling pathway ,Antibody ,Peptides ,Protein Binding ,Signal Transduction - Abstract
We report here the rational design and validation of a peptide inhibitor to the PD-1/PD-L1 interaction as an attempt to develop a viable alternative to current inhibitory antibodies. We demonstrated, by biolayer interferometry and in silico docking simulations, that a PD-L1 peptide mimetic (PL120131) can interfere with the PD-1/PD-L1 interaction by binding to PD-1. We show that PL120131 is capable of inhibiting PD-1 mediated apoptotic signaling pathway and rescuing Jurkat cells and primary lymphocytes from apoptosis. Additionally, we show that PL120131 treatment allows for CTL anti-tumor activity. Furthermore, PL120131 can maintain co-culture survivability and activity of T Cells in a 3D co-culture model better than the anti-PD-1 blocking antibody. Together, the characterization of this PD-1/PD-L1 inhibiting peptide provides insight regarding the ability to inhibit PD-L1 binding while maintaining CTL viability and activity that can further the development of alternatives to antibody based immunotherapies.
- Published
- 2018
37. Thrombospondin-1 regulation of latent TGF-β activation: a therapeutic target for fibrotic disease
- Author
-
Joanne E. Murphy-Ullrich and Mark J. Suto
- Subjects
0301 basic medicine ,Liver Cirrhosis ,Cell ,Disease ,Article ,Diabetes Complications ,Thrombospondin 1 ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Fibrosis ,Transforming Growth Factor beta ,medicine ,Animals ,Humans ,Molecular Biology ,business.industry ,Matricellular protein ,Biological activity ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,business ,Multiple Myeloma ,Peptides ,Transforming growth factor - Abstract
Transforming growth factor-β (TGF-β) is a central player in fibrotic disease. Clinical trials with global inhibitors of TGF-β have been disappointing, suggesting that a more targeted approach is warranted. Conversion of the latent precursor to the biologically active form of TGF-β represents a novel approach to selectively modulating TGF-β in disease, as mechanisms employed to activate latent TGF-β 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-β activation and the use of an antagonist of TSP-1 mediated TGF-β activation in a number of diverse fibrotic diseases. In particular, we will discuss the TSP-1/TGF-β 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-β activity, and in immune dysfunction. As TSP-1 expression is upregulated by factors induced in fibrotic disease, targeting the TSP-1/TGF-β pathway potentially represents a more selective approach to controlling TGF-β activity in disease.
- Published
- 2017
38. Janet A. Houghton, PhD: In Memoriam (1952–2017)
- Author
-
Mark J. Suto
- Subjects
0301 basic medicine ,Cancer Research ,Psychoanalysis ,education ,Cancer ,Cellular level ,medicine.disease ,humanities ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Sociology ,health care economics and organizations - Abstract
[][1] Janet A. Houghton, PhD, a distinguished oncology researcher, passed away on October 5, 2017, at the age of 65 from appendiceal cancer. In a career spanning over 40 years, Dr. Houghton's work deepened scientists' understanding of how to treat cancer at a cellular level.
- Published
- 2018
- Full Text
- View/download PDF
39. Abstract A103: Targeting GLI1-mediated NBS1 transcription overcomes 5-FU resistance
- Author
-
Theresa H Nguyen, Ruowen Zhang, Mark J. Suto, Jinlu Ma, Justin T. Avery, Rebecca J. Boohaker, and Bo Xu
- Subjects
Cancer Research ,integumentary system ,biology ,DNA damage ,Colorectal cancer ,business.industry ,Promoter ,medicine.disease ,Oncology ,FOLFOX ,GLI1 ,Rad50 ,biology.protein ,medicine ,Cancer research ,Gene silencing ,Signal transduction ,business ,medicine.drug - Abstract
Resistance to radiation and chemotherapy in colorectal patients is considered one of the major contributors of refractory disease and disease progression. Dysregulation or aberrant activation of the SHH/GLI1 signaling pathway, common to both colorectal cancer and other cancer subtypes, influences the integrity of the DNA damage response (DDR) pathway, which may impact tumor response to chemo- and radiation therapies. The DDR pathway is initiated in part by the recognition of a DNA lesion by a sensor protein and the recruitment NBS1 to that lesion. Subsequent repair is initiated by the Mre11, Rad50, NBS1 (MRN) complex formation. In this study, we investigated the expression of GLI1 and NBS1 in tissues samples from 188 patients diagnosed with colorectal cancer by immunohistochemistry and analyzed the clinical significance and prognostic relevance of elevated GLI1-NBS1 co-expression. GLI1 expression was positively associated with the NBS1 expression, and high expression of both in the biopsied specimens significantly correlated with poor patient survival. The result of a series of biochemical experiments directly links GLI1 transcriptional activity to the DNA damage repair pathway. First, NBS1 mRNA levels and protein were found to be reduced upon treatment with GANT61, a GLI inhibitor, indicating that GLI activity was required for NBS1 transcription. Second, overexpression of NBS1 in HT29 cells rescued GANT61-induced cell death by measure of cleaved caspase-3, indicating a significant role of induced DNA damage in the mechanism of GANT61-induced cell death in oncogenic GLI1 cancers. Third, this was verified by ChIP analysis of GLI1 binding to the NBS1 promotor region containing a putative GLI binding sequence. The efficacy of GLI1 inhibition as therapeutic strategy was then tested in parallel with 5-Fluorouracil (5-FU), a component of the standard-of-care therapy FOLFOX. In vitro, colorectal cell lines with GLI1-driven NBS1 expression demonstrated strong 5-FU resistance. These cells were re-sensitized to 5-FU by silencing the expression of NBS1 with siRNA, or with combination therapy involving 5-FU and GANT61. This suggests that a first-in class chemotherapeutic strategy may provide additional treatment options for patients demonstrating 5-FU resistance during the course of their chemotherapy. While GANT61 is commonly used to inhibit GLI in vitro, it exhibits poor pharmacokinetic properties and is not a viable drug for the clinic. To that end, we present a novel GLI1 antagonist SR38832, which exhibits a more stable pharmacokinetic profile than GANT61 in xenograft mouse models and lower IC50 in colon cancer cell lines. In a human colorectal cancer xenograft mouse model, SR38832 significantly inhibited both tumor growth and proliferation. Not only has the identification of SR38832 provided an in vivo tool to study GLI oncogenic activity, but SR38832 is also structural lead to be optimized through an iterative medicinal chemistry approach. Citation Format: Ruowen Zhang, Justin Avery, Jinlu Ma, Theresa Nguyen, Mark J Suto, Bo Xu, Rebecca J Boohaker. Targeting GLI1-mediated NBS1 transcription overcomes 5-FU resistance [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A103. doi:10.1158/1535-7163.TARG-19-A103
- Published
- 2019
- Full Text
- View/download PDF
40. Role of Adiponectin in the Metabolic Syndrome: Current Perspectives on its Modulation as a Treatment Strategy
- Author
-
Indira Padmalayam and Mark J. Suto
- Subjects
medicine.medical_specialty ,Adipose tissue ,Peroxisome proliferator-activated receptor ,AMP-Activated Protein Kinases ,Biology ,Insulin resistance ,Metabolic Diseases ,Diabetes mellitus ,Internal medicine ,Drug Discovery ,Nonalcoholic fatty liver disease ,medicine ,Animals ,Humans ,PPAR alpha ,Molecular Targeted Therapy ,Metabolic Syndrome ,Pharmacology ,chemistry.chemical_classification ,Adiponectin ,nutritional and metabolic diseases ,medicine.disease ,Disease Models, Animal ,Endocrinology ,chemistry ,Drug Design ,Metabolic syndrome ,Dyslipidemia - Abstract
Adiponectin, a secretory protein specifically expressed by adipose tissue, has been shown to play a critical role in the maintenance of metabolic homeostasis. A deficiency of adiponectin has been linked to a wide variety of metabolic abnormalities, including obesity and associated disorders such as insulin resistance, hyperglycemia, dyslipidemia, hypertension and nonalcoholic fatty liver disease, collectively referred to as the "metabolic syndrome". Conversely, increased expression of adiponectin corrects these abnormalities, as revealed by the positive metabolic effects observed in genetic over expression studies or by administration of recombinant adiponectin. This has led to widespread interest in its role as a therapeutic target for treatment of a range of metabolic disorders such as diabetes mellitus, obesity, inflammatory and cardiovascular diseases. Various therapeutic approaches targeted at increasing adiponectin levels, or its activity, are being explored. These consist of increasing expression of adiponectin or its receptors by inducers, increasing circulating levels of adiponectin by administering recombinant protein, peptide mimetic approaches, or increasing expression/activity of its downstream effectors such as AMPK or PPAR alpha. Many of these approaches have achieved therapeutic benefits in animal models of metabolic diseases. Despite the profusion of research on adiponectin and ways to modulate it, there are limited number of studies focused on smallmolecule based-therapeutic approaches. In this review, we summarize what is currently known with respect to the therapeutic potential of adiponectin and discuss the challenges in designing small molecule-based therapies.
- Published
- 2013
- Full Text
- View/download PDF
41. Discovery of Clinically Approved Agents That Promote Suppression of Cystic Fibrosis Transmembrane Conductance Regulator Nonsense Mutations
- Author
-
Xiaojiao Xue, David M. Bedwell, Bo Liu, Kim M. Keeling, Martin Mense, Feng Liang, Emily Falk Libby, Marina Mazur, J. Robert Bostwick, Mark J. Suto, Haibo Shang, E. Lucile White, Venkateshwar Mutyam, Ming Du, Jeong S. Hong, Steven M. Rowe, and Lynn Rasmussen
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system diseases ,Cystic Fibrosis ,Nonsense mutation ,Drug Evaluation, Preclinical ,Cystic Fibrosis Transmembrane Conductance Regulator ,Critical Care and Intensive Care Medicine ,Real-Time Polymerase Chain Reaction ,Cystic fibrosis ,Cell Line ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Medicine ,Animals ,Humans ,Luciferase ,Luciferases ,biology ,business.industry ,Translational readthrough ,Original Articles ,Transfection ,medicine.disease ,Molecular biology ,humanities ,Cystic fibrosis transmembrane conductance regulator ,Stop codon ,Rats, Inbred F344 ,030104 developmental biology ,030228 respiratory system ,Codon, Nonsense ,Mutation ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Premature termination codons (PTCs) in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF). Several agents are known to suppress PTCs but are poorly efficacious or toxic.To determine whether there are clinically available agents that elicit translational readthrough and improve CFTR function sufficient to confer therapeutic benefit to patients with CF with PTCs.Two independent screens, firefly luciferase and CFTR-mediated transepithelial chloride conductance assay, were performed on a library of 1,600 clinically approved compounds using fisher rat thyroid cells stably transfected with stop codons. Select agents were further evaluated using secondary screening assays including short circuit current analysis on primary cells from patients with CF. In addition, the effect of CFTR modulators (ivacaftor) was tested in combination with the most efficacious agents.From the primary screen, 48 agents were selected as potentially active. Following confirmatory tests in the transepithelial chloride conductance assay and prioritizing agents based on favorable pharmacologic properties, eight agents were advanced for secondary screening. Ivacaftor significantly increased short circuit current following forskolin stimulation in cells treated with pyranoradine tetraphosphate, potassium p-aminobenzoate, and escin as compared with vehicle control. Escin, an herbal agent, consistently induced readthrough activity as demonstrated by enhanced CFTR expression and function in vitro.Clinically approved drugs identified as potential readthrough agents, in combination with ivacaftor, may induce nonsense suppression to restore therapeutic levels of CFTR function. One or more agents may be suitable to advance to human testing.
- Published
- 2016
42. Identification of Small Molecule Inhibitors of Human Cytochrome c Oxidase That Target Chemoresistant Glioma Cells
- Author
-
Corinne E. Griguer, Mark J. Suto, Maaike Everts, Claudia R. Oliva, Wei Zhang, Lynn Rasmussen, Shannon M. Bailey, Tahireh Markert, Douglas R. Moellering, E. Lucile White, and Larry J. Ross
- Subjects
0301 basic medicine ,Oxidative phosphorylation ,macromolecular substances ,Mitochondrion ,Biology ,Biochemistry ,Respiratory electron transport chain ,Electron Transport Complex IV ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Glioma ,Cell Line, Tumor ,medicine ,Cytochrome c oxidase ,Animals ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Oxidase test ,Cell growth ,Cytochromes c ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,030104 developmental biology ,Tumor progression ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Enzymology - Abstract
The enzyme cytochrome c oxidase (CcO) or complex IV (EC 1.9.3.1) is a large transmembrane protein complex that serves as the last enzyme in the respiratory electron transport chain of eukaryotic mitochondria. CcO promotes the switch from glycolytic to oxidative phosphorylation (OXPHOS) metabolism and has been associated with increased self-renewal characteristics in gliomas. Increased CcO activity in tumors has been associated with tumor progression after chemotherapy failure, and patients with primary glioblastoma multiforme and high tumor CcO activity have worse clinical outcomes than those with low tumor CcO activity. Therefore, CcO is an attractive target for cancer therapy. We report here the characterization of a CcO inhibitor (ADDA 5) that was identified using a high throughput screening paradigm. ADDA 5 demonstrated specificity for CcO, with no inhibition of other mitochondrial complexes or other relevant enzymes, and biochemical characterization showed that this compound is a non-competitive inhibitor of cytochrome c. When tested in cellular assays, ADDA 5 dose-dependently inhibited the proliferation of chemosensitive and chemoresistant glioma cells but did not display toxicity against non-cancer cells. Furthermore, treatment with ADDA 5 led to significant inhibition of tumor growth in flank xenograft mouse models. Importantly, ADDA 5 inhibited CcO activity and blocked cell proliferation and neurosphere formation in cultures of glioma stem cells, the cells implicated in tumor recurrence and resistance to therapy in patients with glioblastoma. In summary, we have identified ADDA 5 as a lead CcO inhibitor for further optimization as a novel approach for the treatment of glioblastoma and related cancers.
- Published
- 2016
43. Discovery of a novel inhibitor of kinesin-like protein KIFC1*
- Author
-
Subramaniam Ananthan, Mark J. Suto, Yimin Wang, Ling Zhai, Rebecca J. Boohaker, Bo Xu, Rongbao Li, Indira Padmalayam, Robert Bostwick, Larry J. Ross, Joseph A. Maddry, Vandana V. Gupta, E. Lucile White, Yonghe Li, Wenyan Lu, Wei Zhang, and Corinne E. Augelli-Szafran
- Subjects
0301 basic medicine ,Genome instability ,Cell Survival ,Kinesins ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,Biochemistry ,Article ,Protein Structure, Secondary ,03 medical and health sciences ,Cell Line, Tumor ,Drug Discovery ,Thiadiazoles ,medicine ,Humans ,Molecular Biology ,Mitosis ,Triple-negative breast cancer ,Dose-Response Relationship, Drug ,Cell Biology ,Cell biology ,030104 developmental biology ,Centrosome ,Cancer cell ,Kinesin ,KIFC1 ,Carcinogenesis ,Protein Binding - Abstract
Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting cancer cells. Therefore, the identification of novel agents that act specifically against cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 μM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast cancer treatment.
- Published
- 2016
44. Process Development and Optimization for Production of a Potassium Ion Channel Blocker, ICA-17043
- Author
-
Kerry Spear, Sripathy Venkatraman, Bingidimi I. Mobele, Cameron Gibb, Brian E. Marron, Grant Andrew Mcnaughton-Smith, Stephen M. Shaw, Mark J. Suto, Luckner Ulysse, and Carl A. Lindmark
- Subjects
Active ingredient ,Hydrolysis ,chemistry.chemical_compound ,Chromatography ,Nitrile ,Chemistry ,Process development ,Organic Chemistry ,Grignard reaction ,Physical and Theoretical Chemistry ,Cyanation ,High-performance liquid chromatography ,Potassium channel - Abstract
A scalable process for the manufacture of a potassium ion channel blocker was developed and optimized. Key features of the process include an optimized Grignard reaction, a direct cyanation of the intermediate trityl alcohol derivative, and an improved nitrile hydrolysis protocol, relative to the original acidic hydrolysis conditions, to generate the crude active pharmaceutical ingredient (API) with >95% HPLC purity. The Grignard and the cyanation reactions could be telescoped, resulting in an improved throughput compared to the original four-step process. An effective recrystallization of the API was also developed and the process scaled up to manufacture multiple batches at the pilot scale.
- Published
- 2012
- Full Text
- View/download PDF
45. Frizzled7 as an emerging target for cancer therapy
- Author
-
Wei Zhang, Mark J. Suto, Yonghe Li, and Taj D. King
- Subjects
Frizzled ,Carcinoma, Hepatocellular ,Beta-catenin ,Breast Neoplasms ,Mice, SCID ,Article ,Mice ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,RNA, Small Interfering ,Receptor ,Wnt Signaling Pathway ,beta Catenin ,biology ,Liver Neoplasms ,Wnt signaling pathway ,Cancer ,LRP6 ,LRP5 ,Cell Biology ,medicine.disease ,Antibodies, Neutralizing ,Frizzled Receptors ,Protein Structure, Tertiary ,Gene Expression Regulation, Neoplastic ,Wnt Proteins ,Gene Knockdown Techniques ,biology.protein ,Cancer research ,Female ,Casein kinase 1 ,Colorectal Neoplasms ,Peptides - Abstract
Wnt proteins are secreted glycoproteins that bind to the N-terminal extra-cellular cysteine-rich domain of the Frizzled (Fzd) receptor family. The Fzd receptors can respond to Wnt proteins in the presence of Wnt co-receptors to activate the canonical and non-canonical Wnt pathways. Recent studies indicated that, among the Fzd family, Fzd7 is the Wnt receptor most commonly upregulated in a variety of cancers including colorectal cancer, hepatocellular carcinoma and triple negative breast cancer. Fzd7 plays an important role in stem cell biology and cancer development and progression. In addition, it has been demonstrated that siRNA knockdown of Fzd7, the anti-Fzd7 antibody or the extracellular peptide of Fzd7 (soluble Fzd7 peptide) displayed anti-cancer activity in vitro and in vivo mainly due to the inhibition of the canonical Wnt signaling pathway. Furthermore, pharmacological inhibition of Fzd7 by small interfering peptides or a small molecule inhibitor suppressed β-catenin-dependent tumor cell growth. Therefore, targeted inhibition of Fzd7 represents a rational and promising new approach for cancer therapy.
- Published
- 2012
- Full Text
- View/download PDF
46. Abstract 5668: The rational design and evaluation of a peptide inhibitor of the PD-1/PD-L1 interaction
- Author
-
Vijaya Sambandam, Mark J. Suto, Rebecca J. Boohaker, and Bo Xu
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,Chemistry ,medicine.medical_treatment ,Immunotherapy ,Jurkat cells ,Immune checkpoint ,03 medical and health sciences ,CTL ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Cytotoxic T cell ,Apoptotic signaling pathway - Abstract
Tumor cells exploit immune checkpoints by expressing checkpoint ligands such as PD-L1, effectively masking them from immune destruction. Programmed cell death- 1 (PD-1), plays a major role in tumor immune escape. The interaction of PD-1/PD-L1 inhibits cytotoxic T lymphocyte (CTL) proliferation, induces apoptosis of tumor-specific T cells, and promotes the resistance of tumor cells to CTL attack. Monoclonal antibodies to these proteins have been developed and approved for use in the clinic. Theoretically, peptide mimics, which have the benefit of lower half-life and significantly smaller size, are able to penetrate into solid tumors and tissues better than antibodies, and can function as inhibitors for the PD1/PD-L1 interaction. Here we report for the first time the rational design and validation of a peptide inhibitor to the PD-1/PD-L1 immune checkpoint as a viable alternative to current inhibitory antibodies. We demonstrated, by biolayer interferometry and in silico docking simulations, that a PD-L1 peptide mimetic (PL120131) can interfere with the PD-1/PD-L1 interaction by binding to PD-1. We established the binding constant for the interaction and PL120131 reduces association of PD-L1 in a dose dependent manner. We show that PL120131 is capable of inhibiting PD-1 mediated apoptotic signaling pathway and rescuing Jurkat cells and primary lymphocytes from apoptosis. Additionally, we show that PL120131 treatment allows for CTL anti-tumor activity. Furthermore, PL120131 can maintain co-culture survivability and activity of T Cells in a 3D co-culture model. Together, the characterization of this PD-1/PD-L1 inhibiting peptide provides insight regarding the ability to inhibit PD-L1 binding while maintaining CTL viability and activity. Understanding the mechanism(s) of action of this peptide has yielded information on the pathways that are drivers of immune evasion in a tumor microenvironment, which may improve and synergize with other forms of immunotherapy, thus increasing efficacy of cancer treatment. Outcomes from this study have provided a path forward to further development of small molecule inhibitors to the PD-1/PD-L1 interaction, and outlines a strategy for the design and development of small molecules to additional cell-surface checkpoint proteins. Citation Format: Rebecca J. Boohaker, Vijaya Sambandam, Mark Suto, Bo Xu. The rational design and evaluation of a peptide inhibitor of the PD-1/PD-L1 interaction [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5668.
- Published
- 2018
- Full Text
- View/download PDF
47. Inhibition of Transforming Growth Factor-β Activation Diminishes Tumor Progression and Osteolytic Bone Disease in Mouse Models of Multiple Myeloma
- Author
-
Joanne E. Murphy-Ullrich, Mark J. Suto, Weiqi Lei, Yang Yang, Huixian Hong, Manuel A. Pallero, and Ailing Lu
- Subjects
0301 basic medicine ,Male ,Osteolysis ,Antineoplastic Agents ,Mice, SCID ,Biology ,Pathology and Forensic Medicine ,Bortezomib ,Thrombospondin 1 ,03 medical and health sciences ,Mice ,Random Allocation ,0302 clinical medicine ,Osteogenesis ,Transforming Growth Factor beta ,hemic and lymphatic diseases ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Multiple myeloma ,Tumor microenvironment ,Interleukin-6 ,Osteoblast ,Cell Differentiation ,Regular Article ,Transforming growth factor beta ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Multiple Myeloma ,Peptides ,medicine.drug ,Transforming growth factor ,Signal Transduction - Abstract
Transforming growth factor (TGF)-β supports multiple myeloma progression and associated osteolytic bone disease. Conversion of latent TGF-β to its biologically active form is a major regulatory node controlling its activity. Thrombospondin1 (TSP1) binds and activates TGF-β. TSP1 is increased in myeloma, and TSP1-TGF-β activation inhibits osteoblast differentiation. We hypothesized that TSP1 regulates TGF-β activity in myeloma and that antagonism of the TSP1-TGF-β axis inhibits myeloma progression. Antagonists (LSKL peptide, SRI31277) derived from the LSKL sequence of latent TGF-β that block TSP1-TGF-β activation were used to determine the role of the TSP1-TGF-β pathway in mouse models of myeloma. TSP1 binds to human myeloma cells and activates TGF-β produced by cultured human and mouse myeloma cell lines. Antagonists delivered via osmotic pump in an intratibial severe combined immunodeficiency CAG myeloma model or in a systemic severe combined immunodeficiency CAG-heparanase model of aggressive myeloma reduced TGF-β signaling (phospho-Smad 2) in bone sections, tumor burden, mouse IL-6, and osteoclasts, increased osteoblast number, and inhibited bone destruction as measured by microcomputed tomography. SRI31277 reduced tumor burden in the immune competent 5TGM1 myeloma model. SRI31277 was as effective as dexamethasone or bortezomib, and SRI31277 combined with bortezomib showed greater tumor reduction than either agent alone. These studies validate TSP1-regulated TGF-β activation as a therapeutic strategy for targeted inhibition of TGF-β in myeloma.
- Published
- 2015
48. High-throughput screening identifies small molecules that enhance the pharmacological effects of oligonucleotides
- Author
-
Mark J. Suto, Emily A. Hull-Ryde, William P. Janzen, Ahu Yuan, Xin Ming, Bing Yang, Joseph A. Maddry, Melissa A. Porter, Rudy L. Juliano, Brian Laing, and Canhong Cao
- Subjects
Endosome ,High-throughput screening ,Oligonucleotides ,Mice, Transgenic ,Biology ,Cell Line ,Small Molecule Libraries ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Chemical Biology and Nucleic Acid Chemistry ,Cell Line, Tumor ,Genetics ,Animals ,Humans ,RNA, Small Interfering ,030304 developmental biology ,0303 health sciences ,Oligonucleotide ,RNA ,Drug Synergism ,Intracellular Membranes ,Oligonucleotides, Antisense ,Molecular biology ,Small molecule ,Cell biology ,High-Throughput Screening Assays ,Cell culture ,030220 oncology & carcinogenesis ,Intracellular - Abstract
The therapeutic use of antisense and siRNA oligonucleotides has been constrained by the limited ability of these membrane-impermeable molecules to reach their intracellular sites of action. We sought to address this problem using small organic molecules to enhance the effects of oligonucleotides by modulating their intracellular trafficking and release from endosomes. A high-throughput screen of multiple small molecule libraries yielded several hits that markedly potentiated the actions of splice switching oligonucleotides in cell culture. These compounds also enhanced the effects of antisense and siRNA oligonucleotides. The hit compounds preferentially caused release of fluorescent oligonucleotides from late endosomes rather than other intracellular compartments. Studies in a transgenic mouse model indicated that these compounds could enhance the in vivo effects of a splice-switching oligonucleotide without causing significant toxicity. These observations suggest that selected small molecule enhancers may eventually be of value in oligonucleotide-based therapeutics.
- Published
- 2015
- Full Text
- View/download PDF
49. Design of a gene family screening library targeting G-protein coupled receptors
- Author
-
Angelo J. Castellino, Peter D. J. Grootenhuis, Bondy Steven Scott, Mark J. Suto, Erin K. Bradley, Graham Beaton, Paul A. Gibbons, Michelle Lamb, and Molecular and Computational Toxicology
- Subjects
Prioritization ,Molecular Structure ,Computer science ,Computational biology ,Ligands ,Bioinformatics ,Computer Graphics and Computer-Aided Design ,Receptors, G-Protein-Coupled ,Drug Design ,Multigene Family ,Materials Chemistry ,Iterative analysis ,Combinatorial Chemistry Techniques ,Gene family ,Physical and Theoretical Chemistry ,Pharmacophore ,Receptor ,Gene ,Spectroscopy ,Gene Library ,G protein-coupled receptor - Abstract
An iterative process for the design of a G-protein coupled receptor (GPCR) gene family screening library has been developed. A key element of this process is the computational generation of pharmacophore descriptors of known GPCR ligands. Subsequent iterative analysis allows prioritization of scaffolds and sub-libraries for inclusion in the library. The final library, which consisted of 13,769 compounds, displayed a 2.6% hit rate when screened against the μ-opioid receptor.
- Published
- 2004
- Full Text
- View/download PDF
50. The design and synthesis of novel orally active inhibitors of AP-1 and NF-κB mediated transcriptional activation. SAR of In vitro and In vivo studies
- Author
-
Moorthy S. S. Palanki, Lynn J. Ransone, Minghuan Ren, Ryuichi Totsuka, Sonal Desai, Arnie Ow, Paul Erdman, Peter W. Tsao, Cheryl Spooner, Brydon L. Bennett, Mark J. Suto, Wataru Toriumi, and Anthony M. Manning
- Subjects
Transcriptional Activation ,Clinical Biochemistry ,Administration, Oral ,Pharmaceutical Science ,Arthritis ,Biochemistry ,Chemical synthesis ,Jurkat Cells ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,Oral administration ,Drug Discovery ,Quinazoline ,medicine ,Animals ,Humans ,Molecular Biology ,Bicyclic molecule ,Organic Chemistry ,NF-kappa B ,NF-κB ,medicine.disease ,Arthritis, Experimental ,In vitro ,Rats ,Transcription Factor AP-1 ,Disease Models, Animal ,chemistry ,Drug Design ,Quinazolines ,Molecular Medicine - Abstract
We have developed novel orally active quinazoline analogues as inhibitors of AP-1 and NF-κB mediated transcriptional activation. Among the derivatives prepared, 1-[2-(2-thienyl)quinazolin-4-ylamino]-3-methyl-3-pyrroline-2,5-dione ( 10 ) showed significant activity in an adjuvant-induced arthritis rat model by reducing the swelling by 65% in the non-injected foot. The synthesis, structure–activity relationship, and in vivo activity are described.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.