41 results on '"Jay H. Kalin"'
Search Results
2. Chemical modulation of Schistosoma mansoni lysine specific demethylase 1 (SmLSD1) induces wide-scale biological and epigenomic changes [version 1; peer review: 2 approved]
- Author
-
Iain W. Chalmers, Christoph Grunau, Josephine Forde-Thomas, Karl F. Hoffmann, Andrea Brancale, Cassandra A. Celatka, Gilda Padalino, Jay H. Kalin, Hugh Y. Rienhoff Jr., Damien Lassalle, and Philip A. Cole
- Subjects
Lysine Specific Demethylase 1 ,epigenetics ,ATAC-seq ,anthelmintics ,eng ,Medicine ,Science - Abstract
Background: Schistosoma mansoni, a parasitic worm species responsible for the neglected tropical disease schistosomiasis, undergoes strict developmental regulation of gene expression that is carefully controlled by both genetic and epigenetic processes. As inhibition of S. mansoni epigenetic machinery components impairs key transitions throughout the parasite’s digenetic lifecycle, a greater understanding of how epi-drugs affect molecular processes in schistosomes could lead to the development of new anthelmintics. Methods: In vitro whole organism assays were used to assess the anti-schistosomal activity of 39 Homo sapiens Lysine Specific Demethylase 1 (HsLSD1) inhibitors on different parasite life cycle stages. Moreover, tissue-specific stains and genomic analysis shed light on the effect of these small molecules on the parasite biology. Results: Amongst this collection of small molecules, compound 33 was the most potent in reducing ex vivo viabilities of schistosomula, juveniles, miracidia and adults. At its sub-lethal concentration to adults (3.13 µM), compound 33 also significantly impacted oviposition, ovarian as well as vitellarian architecture and gonadal/neoblast stem cell proliferation. ATAC-seq analysis of adults demonstrated that compound 33 significantly affected chromatin structure (intragenic regions > intergenic regions), especially in genes differentially expressed in cell populations (e.g., germinal stem cells, hes2+ stem cell progeny, S1 cells and late female germinal cells) associated with these ex vivo phenotypes. KEGG analyses further highlighted that chromatin structure of genes associated with sugar metabolism as well as TGF-beta and Wnt signalling were also significantly perturbed by compound 33 treatment. Conclusions: This work confirms the importance of histone methylation in S. mansoni lifecycle transitions, suggesting that evaluation of LSD1 - targeting epi-drugs may facilitate the search for next-generation anti-schistosomal drugs. The ability of compound 33 to modulate chromatin structure as well as inhibit parasite survival, oviposition and stem cell proliferation warrants further investigations of this compound and its epigenetic target SmLSD1.
- Published
- 2023
- Full Text
- View/download PDF
3. HDAC2 targeting stabilizes the CoREST complex in renal tubular cells and protects against renal ischemia/reperfusion injury
- Author
-
David D. Aufhauser, Paul Hernandez, Seth J. Concors, Ciaran O’Brien, Zhonglin Wang, Douglas R. Murken, Arabinda Samanta, Ulf H. Beier, Lauren Krumeich, Tricia R. Bhatti, Yanfeng Wang, Guanghui Ge, Liqing Wang, Shayan Cheraghlou, Florence F. Wagner, Edward B. Holson, Jay H. Kalin, Philip A. Cole, Wayne W. Hancock, and Matthew H. Levine
- Subjects
Medicine ,Science - Abstract
Abstract Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.
- Published
- 2021
- Full Text
- View/download PDF
4. Targeting the CoREST complex with dual histone deacetylase and demethylase inhibitors
- Author
-
Jay H. Kalin, Muzhou Wu, Andrea V. Gomez, Yun Song, Jayanta Das, Dawn Hayward, Nkosi Adejola, Mingxuan Wu, Izabela Panova, Hye Jin Chung, Edward Kim, Holly J. Roberts, Justin M. Roberts, Polina Prusevich, Jeliazko R. Jeliazkov, Shourya S. Roy Burman, Louise Fairall, Charles Milano, Abdulkerim Eroglu, Charlotte M. Proby, Albena T. Dinkova-Kostova, Wayne W. Hancock, Jeffrey J. Gray, James E. Bradner, Sergio Valente, Antonello Mai, Nicole M. Anders, Michelle A. Rudek, Yong Hu, Byungwoo Ryu, John W. R. Schwabe, Andrea Mattevi, Rhoda M. Alani, and Philip A. Cole
- Subjects
Science - Abstract
Alteration of the epigenetic landscape has been implicated in several disease processes, where targeting histone modifiers may have therapeutic applications. Here the authors report a bifunctional small molecule inhibitor that simultaneously targets the deacetylase (HDAC1) and demethylase (LSD1) activities of the CoREST complex.
- Published
- 2018
- Full Text
- View/download PDF
5. Supplementary Figures 1-7 and Table S1 from Lysine-Specific Demethylase 1 Mediates AKT Activity and Promotes Epithelial-to-Mesenchymal Transition in PIK3CA-Mutant Colorectal Cancer
- Author
-
Heather M. O'Hagan, Gabriel E. Zentner, Philip A. Cole, Jay H. Kalin, Sha Cao, Helai P. Mohammad, Xiaoyu Lu, Ning Ding, Shruthi Sriramkumar, Sudha S. Savant, Robert A. Policastro, and Samuel A. Miller
- Abstract
Supplementary Table S1: Mutational status of common oncogenes in CRC and STAD cell lines. Supplementary Figure S1: Mutant PIK3CA does not regulate LSD1 expression. Supplementary Figure S2: DNase signal is not generally depleted at TSS's, and LSD1 does not reduce global H3K4me2. Supplementary Figure S3: RCOR1 is generally not upregulated in PIK3CA mutant versus WT in cancers arising from non-gastrointestinal tissues, and generally does not correlate with LSD1 expression. Supplementary Figure S4: LSD1 KO reduces cell viability over time in HT29 but not SW480 cells. Supplementary Figure S5: Inhibiting AKT does not reduce Snail protein level in PIK3CA WT or kinase domain mutant cells, and the LSD1-AKT-GSK3b-Snail axis is context dependent. Supplementary Figure S6: LSD1 is upregulated in PIK3CA mutants classified as CRC subtype CMS4, where C2 domain mutations are also most common. Supplementary Figure S7: EGF treatment causes morphological changes in HT29 WT but not LSD1 KO cells.
- Published
- 2023
6. Data from Lysine-Specific Demethylase 1 Mediates AKT Activity and Promotes Epithelial-to-Mesenchymal Transition in PIK3CA-Mutant Colorectal Cancer
- Author
-
Heather M. O'Hagan, Gabriel E. Zentner, Philip A. Cole, Jay H. Kalin, Sha Cao, Helai P. Mohammad, Xiaoyu Lu, Ning Ding, Shruthi Sriramkumar, Sudha S. Savant, Robert A. Policastro, and Samuel A. Miller
- Abstract
Activation of the epithelial-to-mesenchymal transition (EMT) program is a critical mechanism for initiating cancer progression and migration. Colorectal cancers contain many genetic and epigenetic alterations that can contribute to EMT. Mutations activating the PI3K/AKT signaling pathway are observed in >40% of patients with colorectal cancer contributing to increased invasion and metastasis. Little is known about how oncogenic signaling pathways such as PI3K/AKT synergize with chromatin modifiers to activate the EMT program. Lysine-specific demethylase 1 (LSD1) is a chromatin-modifying enzyme that is overexpressed in colorectal cancer and enhances cell migration. In this study, we determine that LSD1 expression is significantly elevated in patients with colorectal cancer with mutation of the catalytic subunit of PI3K, PIK3CA, compared with patients with colorectal cancer with WT PIK3CA. LSD1 enhances activation of the AKT kinase in colorectal cancer cells through a noncatalytic mechanism, acting as a scaffolding protein for the transcription-repressing CoREST complex. In addition, growth of PIK3CA-mutant colorectal cancer cells is uniquely dependent on LSD1. Knockdown or CRISPR knockout of LSD1 blocks AKT-mediated stabilization of the EMT-promoting transcription factor Snail and effectively blocks AKT-mediated EMT and migration. Overall, we uniquely demonstrate that LSD1 mediates AKT activation in response to growth factors and oxidative stress, and LSD1-regulated AKT activity promotes EMT-like characteristics in a subset of PIK3CA-mutant cells.Implications:Our data support the hypothesis that inhibitors targeting the CoREST complex may be clinically effective in patients with colorectal cancer harboring PIK3CA mutations.
- Published
- 2023
7. Supplementary Materials and Methods from Lysine-Specific Demethylase 1 Mediates AKT Activity and Promotes Epithelial-to-Mesenchymal Transition in PIK3CA-Mutant Colorectal Cancer
- Author
-
Heather M. O'Hagan, Gabriel E. Zentner, Philip A. Cole, Jay H. Kalin, Sha Cao, Helai P. Mohammad, Xiaoyu Lu, Ning Ding, Shruthi Sriramkumar, Sudha S. Savant, Robert A. Policastro, and Samuel A. Miller
- Abstract
Contains additional details on bioinformatic analyses, including references for packages used.
- Published
- 2023
8. Lysine-14 acetylation of histone H3 in chromatin confers resistance to the deacetylase and demethylase activities of an epigenetic silencing complex
- Author
-
Mingxuan Wu, Dawn Hayward, Jay H Kalin, Yun Song, John WR Schwabe, and Philip A Cole
- Subjects
histone ,epigenetics ,enzymology ,post-translational modification ,semisynthesis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The core CoREST complex (LHC) contains histone deacetylase HDAC1 and histone demethylase LSD1 held together by the scaffold protein CoREST. Here, we analyze the purified LHC with modified peptide and reconstituted semisynthetic mononucleosome substrates. LHC demethylase activity toward methyl-Lys4 in histone H3 is strongly inhibited by H3 Lys14 acetylation, and this appears to be an intrinsic property of the LSD1 subunit. Moreover, the deacetylase selectivity of LHC unexpectedly shows a marked preference for H3 acetyl-Lys9 versus acetyl-Lys14 in nucleosome substrates but this selectivity is lost with isolated acetyl-Lys H3 protein. This diminished activity of LHC to Lys-14 deacetylation in nucleosomes is not merely due to steric accessibility based on the pattern of sensitivity of the LHC enzymatic complex to hydroxamic acid-mediated inhibition. Overall, these studies have revealed how a single Lys modification can confer a composite of resistance in chromatin to a key epigenetic enzyme complex involved in gene silencing.
- Published
- 2018
- Full Text
- View/download PDF
9. HDAC2 targeting stabilizes the CoREST complex in renal tubular cells and protects against renal ischemia/reperfusion injury
- Author
-
Jay H. Kalin, Seth J. Concors, Lauren N. Krumeich, Tricia R. Bhatti, Wayne W. Hancock, Yanfeng Wang, Zhonglin Wang, Ciaran O’Brien, Guanghui Ge, Douglas R. Murken, Matthew H. Levine, Edward B. Holson, Shayan Cheraghlou, Florence F. Wagner, David D. Aufhauser, Philip A. Cole, Arabinda Samanta, Paul T. Hernandez, Liqing Wang, and Ulf H. Beier
- Subjects
Male ,0301 basic medicine ,Science ,030232 urology & nephrology ,Histone Deacetylase 2 ,Histone Deacetylase 1 ,Pharmacology ,urologic and male genital diseases ,Article ,Kidney Tubules, Proximal ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Animals ,Drug discovery and development ,Enzyme Inhibitors ,Mice, Knockout ,Kidney ,Multidisciplinary ,biology ,Chemistry ,Histone deacetylase 2 ,Endothelins ,HDAC1 ,Acute kidney injury ,Isoenzymes ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Histone ,Reperfusion Injury ,Knockout mouse ,biology.protein ,Medicine ,Female ,Endothelin receptor ,Co-Repressor Proteins ,Gene Deletion - Abstract
Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.
- Published
- 2021
10. Chemical modulation of Schistosoma mansoni lysine specific demethylase 1 (SmLSD1) induces wide-scale biological and epigenomic changes
- Author
-
Gilda Padalino, Cassandra A. Celatka, Hugh Y. Rienhoff Jr., Jay H. Kalin, Philip A. Cole, Damien Lassalle, Josephine Forde-Thomas, Iain W. Chalmers, Andrea Brancale, Christoph Grunau, and Karl F. Hoffmann
- Subjects
Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology - Abstract
Background: Schistosoma mansoni, a parasitic worm species responsible for the neglected tropical disease schistosomiasis, undergoes strict developmental regulation of gene expression that is carefully controlled by both genetic and epigenetic processes. As inhibition of S. mansoni epigenetic machinery components impairs key transitions throughout the parasite’s digenetic lifecycle, a greater understanding of how epi-drugs affect molecular processes in schistosomes could lead to the development of new anthelmintics. Methods: In vitro whole organism assays were used to assess the anti-schistosomal activity of 39 Homo sapiens Lysine Specific Demethylase 1 (HsLSD1) inhibitors on different parasite life cycle stages. Moreover, tissue-specific stains and genomic analysis shed light on the effect of these small molecules on the parasite biology. Results: Amongst this collection of small molecules, compound 33 was the most potent in reducing ex vivo viabilities of schistosomula, juveniles, miracidia and adults. At its sub-lethal concentration to adults (3.13 µM), compound 33 also significantly impacted oviposition, ovarian as well as vitellarian architecture and gonadal/neoblast stem cell proliferation. ATAC-seq analysis of adults demonstrated that compound 33 significantly affected chromatin structure (intragenic regions > intergenic regions), especially in genes differentially expressed in cell populations (e.g., germinal stem cells, hes2+ stem cell progeny, S1 cells and late female germinal cells) associated with these ex vivo phenotypes. KEGG analyses further highlighted that chromatin structure of genes associated with sugar metabolism as well as TGF-beta and Wnt signalling were also significantly perturbed by compound 33 treatment. Conclusions: This work confirms the importance of histone methylation in S. mansoni lifecycle transitions, suggesting that evaluation of LSD1 - targeting epi-drugs may facilitate the search for next-generation anti-schistosomal drugs. The ability of compound 33 to modulate chromatin structure as well as inhibit parasite survival, oviposition and stem cell proliferation warrants further investigations of this compound and its epigenetic target SmLSD1.
- Published
- 2023
11. Lysine-Specific Demethylase 1 Mediates AKT Activity and Promotes Epithelial-to-Mesenchymal Transition in PIK3CA-Mutant Colorectal Cancer
- Author
-
Shruthi Sriramkumar, Gabriel E. Zentner, Helai P. Mohammad, Philip A. Cole, Robert A. Policastro, Samuel A. Miller, Sudha S. Savant, Sha Cao, Heather M. O'Hagan, Jay H. Kalin, Xiaoyu Lu, and Ning Ding
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,Chemistry ,Akt/PKB signaling pathway ,Colorectal cancer ,Cancer ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Epithelial–mesenchymal transition ,Molecular Biology ,Transcription factor ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Activation of the epithelial-to-mesenchymal transition (EMT) program is a critical mechanism for initiating cancer progression and migration. Colorectal cancers contain many genetic and epigenetic alterations that can contribute to EMT. Mutations activating the PI3K/AKT signaling pathway are observed in >40% of patients with colorectal cancer contributing to increased invasion and metastasis. Little is known about how oncogenic signaling pathways such as PI3K/AKT synergize with chromatin modifiers to activate the EMT program. Lysine-specific demethylase 1 (LSD1) is a chromatin-modifying enzyme that is overexpressed in colorectal cancer and enhances cell migration. In this study, we determine that LSD1 expression is significantly elevated in patients with colorectal cancer with mutation of the catalytic subunit of PI3K, PIK3CA, compared with patients with colorectal cancer with WT PIK3CA. LSD1 enhances activation of the AKT kinase in colorectal cancer cells through a noncatalytic mechanism, acting as a scaffolding protein for the transcription-repressing CoREST complex. In addition, growth of PIK3CA-mutant colorectal cancer cells is uniquely dependent on LSD1. Knockdown or CRISPR knockout of LSD1 blocks AKT-mediated stabilization of the EMT-promoting transcription factor Snail and effectively blocks AKT-mediated EMT and migration. Overall, we uniquely demonstrate that LSD1 mediates AKT activation in response to growth factors and oxidative stress, and LSD1-regulated AKT activity promotes EMT-like characteristics in a subset of PIK3CA-mutant cells. Implications: Our data support the hypothesis that inhibitors targeting the CoREST complex may be clinically effective in patients with colorectal cancer harboring PIK3CA mutations.
- Published
- 2020
12. Development and characterization of rabbit monoclonal antibodies that recognize human spermine oxidase and application to immunohistochemistry of human cancer tissues
- Author
-
Armand W. J. W. Tepper, Gerald Chu, Vincent N. A. Klaren, Jay H. Kalin, Patricia Molina-Ortiz, and Antonietta Impagliazzo
- Subjects
Male ,Oxidoreductases Acting on CH-NH Group Donors ,Multidisciplinary ,Antineoplastic Agents, Immunological ,Neoplasms ,Antibodies, Monoclonal ,Humans ,Spermine ,Immunohistochemistry - Abstract
The enzyme spermine oxidase (SMOX) is involved in polyamine catabolism and converts spermine to spermidine. The enzymatic reaction generates reactive hydrogen peroxide and aldehydes as by-products that can damage DNA and other biomolecules. Increased expression of SMOX is frequently found in lung, prostate, colon, stomach and liver cancer models, and the enzyme also appears to play a role in neuronal dysfunction and vascular retinopathy. Because of growing evidence that links SMOX activity with DNA damage, inflammation, and carcinogenesis, the enzyme has come into view as a potential drug target. A major challenge in cancer research is the lack of characterization of antibodies used for identification of target proteins. To overcome this limitation, we generated a panel of high-affinity rabbit monoclonal antibodies against various SMOX epitopes and selected antibodies for use in immunoblotting, SMOX quantification assays, immunofluorescence microscopy and immunohistochemistry. Immunohistochemistry analysis with the antibody SMAB10 in normal and transformed tissues confirms that SMOX is upregulated in several different cancers. Together, the panel of antibodies generated herein adds to the toolbox of high-quality reagents to study SMOX biology and to facilitate SMOX drug development.
- Published
- 2021
13. The CoREST Repressor Complex Mediates Phenotype Switching and Therapy Resistance in Melanoma
- Author
-
Muzhou Wu, Kevin H. Kuang, Philip A. Cole, Frederick Gibson, Jay H. Kalin, Ailish Hanly, Marianne Collard, Matthew W. Cole, Sarah E. Nocco, Adam Labadorf, Rhoda M. Alani, Amy Xiao, and Filisia Agus
- Subjects
Melanoma ,medicine.medical_treatment ,Repressor ,Context (language use) ,Biology ,medicine.disease ,Phenotype ,Targeted therapy ,Mediator ,Cancer research ,medicine ,Epigenetics ,neoplasms ,Reprogramming - Abstract
Virtually all patients with BRAF-mutant melanoma develop resistance to MAPK inhibitors largely through non-mutational events1,2. Although the epigenetic landscape has been shown to be altered in therapy-resistant melanomas and other cancers3,4, a specific targetable epigenetic mechanism regulating treatment resistance has not been validated to date. Here we evaluate the CoREST repressor complex and the novel inhibitor, corin5, within the context of melanoma phenotype plasticity and therapeutic resistance in order to define epigenetic mechanisms underlying these processes. We find that CoREST is a critical mediator of the major distinct melanoma phenotypes and that corin treatment of melanoma cells leads to phenotype reprogramming. We further demonstrate that treatment of BRAF inhibitor (BRAFi)-resistant melanomas with corin leads to resensitization of tumor cells to BRAFi. Among the transcriptional targets of CoREST in melanoma are the dual-specificity phosphatases (DUSPs). DUSP1 is shown to be consistently downregulated in BRAFi-resistant melanomas which can be reversed by corin treatment, thereby leading to downstream inhibition of p38 MAPK activity and resensitization of resistant cells to targeted BRAFi therapies. These findings identify the CoREST repressor complex as a central mediator of melanoma phenotype plasticity and resistance to targeted therapy and suggest that CoREST inhibitors may prove beneficial to patients with BRAF-mutant melanomas who have acquired BRAFi-resistance.
- Published
- 2020
14. Schistosoma mansoni lysine specific demethylase 1 (SmLSD1) is a druggable target involved in parasite survival, oviposition and stem cell proliferation
- Author
-
H. Y. Rienhoff, D. Lassalle, Iain W. Chalmers, C. A. Celatka, Karl F. Hoffmann, Philip A. Cole, Christoph Grunau, Gilda Padalino, Jay H. Kalin, and Andrea Brancale
- Subjects
Regulation of gene expression ,biology ,Schistosomiasis ,biology.organism_classification ,medicine.disease ,Cell biology ,Histone ,parasitic diseases ,Histone methylation ,biology.protein ,medicine ,Demethylase ,Schistosoma mansoni ,Epigenetics ,Stem cell - Abstract
Schistosomiasis is a chronically-debilitating neglected tropical disease (NTD) that predominantly affects people living in resource-poor communities of tropical and subtropical countries. Schistosoma mansoni, one of three species responsible for most human infections, undergoes strict developmental regulation of gene expression that is carefully controlled by both genetic- and epigenetic- processes. As inhibition of S. mansoni epigenetic machinery components has been shown to impair key transitions throughout the parasite’s digenetic lifecycle, this knowledge is currently fuelling the search for new epi-drug - based anthelmintics.In this study, the anti-schistosomal activity of 39 re-purposed Homo sapiens Lysine Specific Demethylase 1 (HsLSD1) inhibitors was investigated on key life cycle stages associated with both definitive (schistosomula, juvenile worms, sexually-mature adults) and intermediate host (miracidia) infection. The most active compound (compound 33; e.g. schistosomula phenotype EC50 = 4.370 µM; adult worm motility EC50 = 2.137 µM) was subsequently used to provide further insight into the critical role of S. mansoni lysine specific demethylase 1 (SmLSD1) in adult worm oviposition and stem cell proliferation. Here, compound 33 treatment of adult schistosomes led to significant defects in egg production, intra-egg vitellocyte/ovum packaging and gonadal/neoblast stem cell proliferation. A greater abundance of H3K4me2 marks accompanied these phenotypes and supported specific inhibition of SmLSD1 in adult schistosomes by compound 33. In silico screening indicated that compound 33 likely inhibits SmLSD1 activity by covalently reacting with the FAD cofactor.This work suggests that evaluation of HsLSD1 - targeting epi-drugs could have utility in the search for next-generation anti-schistosomals. The ability of compound 33 to inhibit parasite survival, oviposition, H3K4me2 demethylation and stem cell proliferation warrants further investigations of this compound and its epigenetic target. This data further highlights the importance of histone methylation in S. mansoni lifecycle transitions.Author summaryAffecting over 200 million people, schistosomiasis is a chronic disease caused by the parasitic worm Schistosoma mansoni. The frontline drug for schistosomiasis treatment is praziquantel. Owing to the concern surrounding praziquantel insensitivity or resistance developing, current research is directed towards the identification of novel drugs. We have focused our search for compounds that affect essential aspects of schistosome biology including parasite movement, fertility, cell proliferation and survival. Since all of these functions are potentially influenced by epigenetic regulation of gene expression, we investigated the activity of compounds that alter histone methylation status. In this report, we show that S. mansoni Lysine Specific Demethylase 1 (SmLSD1), a histone demethylase, is critical to miracidia-to-sporocyst transitioning, adult worm motility, egg production and parasite survival. Inhibition of SmLSD1 with compounds developed to inhibit the human paralog show promising potential as novel anti-schistosomal agents.
- Published
- 2020
15. Re-programing chromatin with a bifunctional LSD1/HDAC inhibitor induces therapeutic differentiation in DIPG
- Author
-
Shawn M. Gillespie, Mariella G. Filbin, Mirhee Kim, Sanda Alexandrescu, Rhoda M. Alani, Muzhou Wu, Barry M. Zee, Bradley E. Bernstein, Jayanta Das, Jay H. Kalin, Dennis M. Bonal, Mario Andres Blanco, Yang Shi, Jamie N. Anastas, Mario L. Suvà, Philip A. Cole, Sarah E. Nocco, Stefanie Giera, Quang-Dé Nguyen, Todd R. Golub, and Robyn Guo
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,Cellular differentiation ,Antineoplastic Agents ,Histone Deacetylases ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Pons ,Animals ,Brain Stem Neoplasms ,Humans ,RNA-Seq ,Epigenetics ,Histone Demethylases ,biology ,Chemistry ,Cell Differentiation ,KDM1A ,Glioma ,Cell Biology ,DNA Methylation ,Xenograft Model Antitumor Assays ,Pediatric cancer ,Chromatin ,Gene Expression Regulation, Neoplastic ,Histone Code ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Histone ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Demethylase ,Female ,Histone deacetylase - Abstract
Summary H3K27M mutations resulting in epigenetic dysfunction are frequently observed in diffuse intrinsic pontine glioma (DIPGs), an incurable pediatric cancer. We conduct a CRISPR screen revealing that knockout of KDM1A encoding lysine-specific demethylase 1 (LSD1) sensitizes DIPG cells to histone deacetylase (HDAC) inhibitors. Consistently, Corin, a bifunctional inhibitor of HDACs and LSD1, potently inhibits DIPG growth in vitro and in xenografts. Mechanistically, Corin increases H3K27me3 levels suppressed by H3K27M histones, and simultaneously increases HDAC-targeted H3K27ac and LSD1-targeted H3K4me1 at differentiation-associated genes. Corin treatment induces cell death, cell-cycle arrest, and a cellular differentiation phenotype and drives transcriptional changes correlating with increased survival time in DIPG patients. These data suggest a strategy for treating DIPG by simultaneously inhibiting LSD1 and HDACs.
- Published
- 2020
16. Inhibiting the coregulator CoREST impairs Foxp3(+) Treg function and promotes antitumor immunity
- Author
-
Eros Di Giorgio, Rongxiang Han, Philip A. Cole, Tatiana Akimova, Liqing Wang, Ulf H. Beier, Wayne W. Hancock, Matteo Trevisanut, Jay H. Kalin, and Yan Xiong
- Subjects
0301 basic medicine ,Scaffold protein ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Biology ,T-Lymphocytes, Regulatory ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Gene expression ,medicine ,Histone deacetylase 2 ,FOXP3 ,hemic and immune systems ,General Medicine ,HDAC1 ,Cell biology ,RCOR1 ,030104 developmental biology ,030220 oncology & carcinogenesis ,Transplantation Tolerance ,Co-Repressor Proteins ,Research Article ,Transcription Factors - Abstract
Foxp3(+) Tregs are key to immune homeostasis, but the contributions of various large, multiprotein complexes that regulate gene expression remain unexplored. We analyzed the role in Tregs of the evolutionarily conserved CoREST complex, consisting of a scaffolding protein, Rcor1 or Rcor2, plus Hdac1 or Hdac2 and Lsd1 enzymes. Rcor1, Rcor2, and Lsd1 were physically associated with Foxp3, and mice with conditional deletion of Rcor1 in Foxp3(+) Tregs had decreased proportions of Tregs in peripheral lymphoid tissues and increased Treg expression of IL-2 and IFN-γ compared with what was found in WT cells. Mice with conditional deletion of the gene encoding Rcor1 in their Tregs had reduced suppression of homeostatic proliferation, inability to maintain long-term allograft survival despite costimulation blockade, and enhanced antitumor immunity in syngeneic models. Comparable findings were seen in WT mice treated with CoREST complex bivalent inhibitors, which also altered the phenotype of human Tregs and impaired their suppressive function. Our data point to the potential for therapeutic modulation of Treg functions by pharmacologic targeting of enzymatic components of the CoREST complex and contribute to an understanding of the biochemical and molecular mechanisms by which Foxp3 represses large gene sets and maintains the unique properties of this key immune cell.
- Published
- 2020
17. Site-Specific Protein Labeling with N-Hydroxysuccinimide-Esters and the Analysis of Ubiquitin Ligase Mechanisms
- Author
-
Philip A. Cole, Hanjie Jiang, Zan Chen, Jay H. Kalin, and Daniel R. Dempsey
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Biotin ,Succinimides ,WWP2 ,Saccharomyces cerevisiae ,Plasma protein binding ,Spodoptera ,01 natural sciences ,Biochemistry ,Article ,Catalysis ,Molecularity ,03 medical and health sciences ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Protein structure ,Ubiquitin ,Escherichia coli ,Animals ,Humans ,Cysteine ,Uracil-DNA Glycosidase ,Fluorescent Dyes ,Glutathione Transferase ,biology ,Rhodamines ,010405 organic chemistry ,PTEN Phosphohydrolase ,Ubiquitination ,Esters ,General Chemistry ,Fluoresceins ,0104 chemical sciences ,Ubiquitin ligase ,030104 developmental biology ,N-Hydroxysuccinimide ,chemistry ,biology.protein ,Function (biology) ,Protein Binding - Abstract
N-hydroxysuccinimide (NHS)-esters are widely used to label proteins non-selectively on free amino groups. Such broad labeling can be disadvantageous because it can interfere with protein structure or function and because stoichiometry is poorly controlled. Here we describe a simple method to transform NHS-esters into site-specific protein labeling on N-terminal Cys residues. MESNA addition converts NHS-esters to chemoselective thioesters for N-Cys modification. This labeling strategy was applied to clarify mechanistic features of the ubiquitin E3 ligase WWP2 including its interaction with one of its substrate, the tumor suppressor PTEN, as well as its autoubiquitination molecularity. We propose that this convenient protein labeling strategy will allow for an expanded application of NHS-esters in biochemical investigation.
- Published
- 2018
18. Investigation into the use of histone deacetylase inhibitor MS-275 as a topical agent for the prevention and treatment of cutaneous squamous cell carcinoma in an SKH-1 hairless mouse model
- Author
-
W. David Holtzclaw, Jay H. Kalin, Abdulkerim Eroglu, Albena T. Dinkova-Kostova, Hua Liu, Charlotte M. Proby, Irene M. Leigh, Jed W. Fahey, and Philip A. Cole
- Subjects
Neoplasms, Radiation-Induced ,Skin Neoplasms ,Light ,Carcinogenesis ,Pyridines ,Administration, Topical ,Cell ,Cancer Treatment ,Gene Expression ,medicine.disease_cause ,Biochemistry ,Histones ,030207 dermatology & venereal diseases ,Mice ,0302 clinical medicine ,Medicine and Health Sciences ,Post-Translational Modification ,Multidisciplinary ,integumentary system ,Chromosome Biology ,Physics ,Chromatin Modification ,Histone deacetylase inhibitor ,Chemical Reactions ,Squamous Cell Carcinomas ,Acetylation ,Histone Modification ,Animal Models ,Chromatin ,3. Good health ,Tumor Burden ,Physical sciences ,Gene Expression Regulation, Neoplastic ,Chemistry ,medicine.anatomical_structure ,Oncology ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Benzamides ,Carcinoma, Squamous Cell ,Medicine ,Epigenetics ,Keratinocyte ,Research Article ,Ultraviolet radiation ,Cyclin-Dependent Kinase Inhibitor p21 ,medicine.drug_class ,Cell Survival ,Science ,Histone Acetylation ,Mouse Models ,Research and Analysis Methods ,Carcinomas ,03 medical and health sciences ,Model Organisms ,Electromagnetic radiation ,Cell Line, Tumor ,DNA-binding proteins ,Carcinoma ,medicine ,Genetics ,Animals ,Humans ,Cell Proliferation ,Mice, Hairless ,Biology and life sciences ,business.industry ,Proteins ,Cancers and Neoplasms ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Hairless ,Histone Deacetylase Inhibitors ,Cancer research ,Animal Studies ,Histone deacetylase ,business ,Ultraviolet B - Abstract
Cutaneous squamous cell carcinomas are a common form of highly mutated keratinocyte skin cancers that are of particular concern in immunocompromised patients. Here we report on the efficacy of topically applied MS-275, a clinically used histone deacetylase inhibitor, for the treatment and management of this disease. At 2 mg/kg, MS-275 significantly decreased tumor burden in an SKH-1 hairless mouse model of UVB radiation-induced skin carcinogenesis. MS-275 was cell permeable as a topical formulation and induced histone acetylation changes in mouse tumor tissue. MS-275 was also effective at inhibiting the proliferation of patient derived cutaneous squamous cell carcinoma lines and was particularly potent toward cells isolated from a regional metastasis on an immunocompromised individual. Our findings support the use of alternative routes of administration for histone deacetylase inhibitors in the treatment of high-risk squamous cell carcinoma which may ultimately lead to more precise delivery and reduced systemic toxicity.
- Published
- 2019
19. Exploration of the labeling of [11C]tubastatin A at the hydroxamic acid site with [11C]carbon monoxide
- Author
-
Shuiyu Lu, Lisheng Cai, Victor W. Pike, Yi Zhang, Alan P. Kozikowski, and Jay H. Kalin
- Subjects
chemistry.chemical_classification ,Hydroxamic acid ,Base (chemistry) ,010405 organic chemistry ,Aryl ,Organic Chemistry ,Iodide ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Medicinal chemistry ,0104 chemical sciences ,Analytical Chemistry ,chemistry.chemical_compound ,Hydroxylamine ,chemistry ,Drug Discovery ,Organic chemistry ,Radiology, Nuclear Medicine and imaging ,Carbonylation ,Spectroscopy ,Phosphazene ,Carbon monoxide - Abstract
We aimed to label tubastatin A (1) with carbon-11 (t1/2 = 20.4 min) in the hydroxamic acid site to provide a potential radiotracer for imaging histone deacetylase 6 in vivo with positron emission tomography. Initial attempts at a one-pot Pd-mediated insertion of [(11)C]carbon monoxide between the aryl iodide (2) and hydroxylamine gave low radiochemical yields (
- Published
- 2015
20. GENE-22. RE-PROGRAMING CHROMATIN WITH A BIFUNCTIONAL LSD1/HDAC INHIBITOR INDUCES THERAPEUTIC DIFFERENTIATION IN DIPG
- Author
-
Andres Blanco, Barry Zee, Mirhee Kim, Jamie N. Anastas, Mariella G. Filbin, Todd R. Golub, Philip A. Cole, Jayanta Das, Jay H. Kalin, and Yang Shi
- Subjects
Cancer Research ,Mutation ,Programmed cell death ,biology ,Chemistry ,medicine.disease_cause ,Chromatin ,Abstracts ,Histone ,Oncology ,medicine ,biology.protein ,Neuron differentiation ,Cancer research ,Neurology (clinical) ,Epigenetics ,Histone deacetylase ,Gene - Abstract
Diffuse intrinsic high grade glioma (DIPG) is an almost universally fatal tumor of childhood characterized by epigenetic dysregulation driven by somatic H3.3/H3.1 K27M mutations observed in >80% of tumors. We conducted a chromatin-focused CRISPR screen and identified a novel strategy to inhibit the growth of patient-derived DIPG cells by co-targeting lysine specific demethylase 1 (LSD1) and histone deacetylases (HDACs). Consistent with the genetic data, we demonstrate that a bifunctional inhibitor of HDACs and LSD1, Corin, inhibits the growth of DIPG cells both in vitro and in orthotopic xenografts. Mechanistically, co-targeting LSD1 and HDACs with Corin synergistically alters the levels of histone modifications in DIPG, rescuing H3K27me3 levels suppressed by the dominant negative effects of K27M mutant histones and inducing simultaneous increases in both HDAC-targeted H3K27ac and LSD1-targeted H3K4me1 at thousands of genomic locations. Coincident with these chromatin changes, we observe robust transcriptional changes in DIPG cells, including the repression of cell cycle-related genes and the activation of neuronal differentiation genes. Consistently, phenotypic assays reveal that Corin reduces S-phase and Ki67+ proliferating cells, induces a neuronal-like morphology, alters the expression of stem and differentiation markers, and increases DIPG cell death. Finally, analyses of patient DIPG expression datasets indicate that Corin-dependent transcriptional signatures are overrepresented in normal brain compared to DIPG tumors and correlate with increased patient survival time. Together, these data reveal that co-inhibiting LSD1 and HDACs synergistically reduces DIPG growth by re-programming the chromatin landscape to activate a latent differentiation response, and suggest a strategy of co-inhibiting LSD1 and HDACs with Corin to treat DIPG.
- Published
- 2018
21. Author response: Lysine-14 acetylation of histone H3 in chromatin confers resistance to the deacetylase and demethylase activities of an epigenetic silencing complex
- Author
-
Mingxuan Wu, Dawn Hayward, Jay H Kalin, Yun Song, John WR Schwabe, and Philip A Cole
- Published
- 2018
22. Analysis of CoREST Complex‐Chromatin Interactions with Chemical Tools
- Author
-
John W.R. Schwabe, Philip A. Cole, Dawn Hayward, Mingxuan Wu, Jay H. Kalin, and Yun Song
- Subjects
Chemistry ,Genetics ,Molecular Biology ,Biochemistry ,Biotechnology ,Chromatin ,Cell biology - Published
- 2018
23. An Fc Domain Protein–Small Molecule Conjugate as an Enhanced Immunomodulator
- Author
-
Jay H. Kalin, Meng-Jung Chiang, Mohammed N. Amin, Young Hoon Ahn, Samuel L. Collins, Philip A. Cole, Martin S. Taylor, Jonathan D. Powell, David M. Bolduc, Lai-Xi Wang, Maureen R. Horton, Yee Chan-Li, Adam T. Waickman, Marc A. Holbert, John P. Giddens, Po Yuan Hsiao, and Daniel J. Leahy
- Subjects
CD4-Positive T-Lymphocytes ,Models, Molecular ,Agonist ,Adenosine ,Immunoconjugates ,Protein Conformation ,medicine.drug_class ,Immunoglobulin Fc ,Fc receptor ,Biochemistry ,Catalysis ,Mice ,Colloid and Surface Chemistry ,Protein structure ,Phenethylamines ,medicine ,Animals ,Immunologic Factors ,Receptor ,biology ,Chemistry ,Communication ,Immunoglobulin Fc Fragments ,General Chemistry ,Small molecule ,Cell biology ,biology.protein ,medicine.drug - Abstract
Proteins as well as small molecules have demonstrated success as therapeutic agents, but their pharmacologic properties sometimes fall short against particular drug targets. Although the adenosine 2a receptor (A(2A)R) has been identified as a promising target for immunotherapy, small molecule A(2A)R agonists have suffered from short pharmacokinetic half-lives and the potential for toxicity by modulating nonimmune pathways. To overcome these limitations, we have tethered the A(2A)R agonist CGS-21680 to the immunoglobulin Fc domain using expressed protein ligation with Sf9 cell secreted protein. The protein small molecule conjugate Fc-CGS retained potent Fc receptor and A(2A)R interactions and showed superior properties as a therapeutic for the treatment of a mouse model of autoimmune pneumonitis. This approach may provide a general strategy for optimizing small molecule therapeutics.
- Published
- 2014
24. 941 Epigenetic manipulation of epidermal cells leads to enhanced cellular migration without an increase in cell proliferation
- Author
-
M. Kida, Muzhou Wu, Philip A. Cole, Jay H. Kalin, Rhoda M. Alani, and Vincent Falanga
- Subjects
Chemistry ,Cell migration ,Cell Biology ,Dermatology ,Epigenetics ,Molecular Biology ,Biochemistry ,Cell biology - Published
- 2019
25. Development and Therapeutic Implications of Selective Histone Deacetylase 6 Inhibitors
- Author
-
Jay H. Kalin and Joel A. Bergman
- Subjects
Histone Deacetylase Inhibitors ,Drug Discovery ,Humans ,Molecular Medicine ,Disease classification ,Disease ,Computational biology ,HDAC6 ,Biology ,Histone Deacetylase 6 ,Bioinformatics ,Histone Deacetylases - Abstract
This Perspective provides an in depth look at the numerous disease states in which histone deacetylase 6 (HDAC6) has been implicated. The physiological pathways, protein-protein interactions, and non-histone substrates relating to different pathological conditions are discussed with regard to HDAC6. Furthermore, the compounds and methods used to modulate HDAC6 activity are profiled. The latter half of this Perspective analyzes reported HDAC6 selective inhibitors in terms of structure, potency, and selectivity over the other HDAC isoforms with the intent of providing a comprehensive overview of the molecular tools available. Potential obstacles and future directions of HDAC6 research are also presented.
- Published
- 2013
26. An Optimized RAD51 Inhibitor That Disrupts Homologous Recombination without Requiring Michael Acceptor Reactivity
- Author
-
Anna Zelivianskaia, Jay H. Kalin, Megan Wu, Michal Pawlowski, Philip P. Connell, Alan P. Kozikowski, and Brian Budke
- Subjects
DNA Repair ,Morpholines ,RAD51 ,Article ,Maleimides ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Humans ,Pyrroles ,Homologous Recombination ,Replication protein A ,Maleimide ,Binding Sites ,DNA replication ,DNA ,HEK293 Cells ,Biochemistry ,chemistry ,Cancer cell ,Michael reaction ,Molecular Medicine ,Rad51 Recombinase ,Homologous recombination ,Protein Binding - Abstract
Homologous recombination (HR) is an essential process in cells that provides repair of DNA double-strand breaks and lesions that block DNA replication. RAD51 is an evolutionarily conserved protein that is central to HR. Overexpression of RAD51 protein is common in cancer cells and represents a potential therapeutic target in oncology. We previously described a chemical inhibitor of RAD51, called RI-1 (referred to as compound 1 in this report). The chloromaleimide group of this compound is thought to act as a Michael acceptor and react with the thiol group on C319 of RAD51, using a conjugate addition-elimination mechanism. In order to reduce the likelihood of off-target effects and to improve compound stability in biological systems, we developed an analogue of compound 1 that lacks maleimide-based reactivity but retains RAD51 inhibitory activity. This compound, 1-(3,4-dichlorophenyl)-3-(4-methoxyphenyl)-4-morpholino-1H-pyrrole-2,5-dione, named RI-2 (referred to as compound 7a in this report), appears to bind reversibly to the same site on the RAD51 protein as does compound 1. Like compound 1, compound 7a specifically inhibits HR repair in human cells.
- Published
- 2012
27. An Fc-Small Molecule Conjugate for Targeted Inhibition of the Adenosine 2A Receptor
- Author
-
Jay H. Kalin, Philip A. Cole, Meng Jung Chiang, Kathleen L. Gabrielson, Jonathan D. Powell, Polina Sysa-Shah, Im Hong Sun, Lai-Xi Wang, John P. Giddens, Ying-Chun Lo, Po Yuan Hsiao, and Mohammed N. Amin
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Receptor, Adenosine A2A ,Adenosine A2A receptor ,Vaccinia virus ,Pharmacology ,Biochemistry ,Article ,03 medical and health sciences ,Immunoglobulin Fab Fragments ,Mice ,Neonatal Fc receptor ,medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,Respiratory Tract Infections ,Mice, Knockout ,biology ,Molecular Structure ,Chemistry ,Triazines ,Organic Chemistry ,Antagonist ,Triazoles ,Small molecule ,Adenosine receptor ,Adenosine ,Adenosine A2 Receptor Antagonists ,Mice, Inbred C57BL ,030104 developmental biology ,biology.protein ,Molecular Medicine ,Female ,Antibody ,medicine.drug - Abstract
The adenosine A2A receptor (A2AR) is expressed in immune cells, as well as brain and heart tissue, and has been intensively studied as a therapeutic target for multiple disease indications. Inhibitors of the A2AR have the potential for stimulating immune response, which could be valuable for cancer immune surveillance and mounting a response against pathogens. One well-established potent and selective small molecule A2AR antagonist, ZM-241385 (ZM), has a short pharmacokinetic half-life and the potential for systemic toxicity due to A2AR effects in the brain and the heart. In this study, we designed an analogue of ZM and tethered it to the Fc domain of the immunoglobulin IgG3 by using expressed protein ligation. The resulting protein–small molecule conjugate, Fc–ZM, retained high affinity for two Fc receptors: FcγRI and the neonatal Fc receptor, FcRn. In addition, Fc–ZM was a potent A2AR antagonist, as measured by a cell-based cAMP assay. Cell-based assays also revealed that Fc–ZM could stimulate interferon γ production in splenocytes in a fashion that was dependent on the presence of A2AR. We found that Fc–ZM, compared with the small molecule ZM, was a superior A2AR antagonist in mice, consistent with the possibility that Fc attachment can improve pharmacokinetic and/or pharmacodynamic properties of the small molecule.
- Published
- 2016
28. Second-Generation Histone Deacetylase 6 Inhibitors Enhance the Immunosuppressive Effects of Foxp3+ T-Regulatory Cells
- Author
-
Kyle V. Butler, Jay H. Kalin, Wayne W. Hancock, Tatiana Akimova, and Alan P. Kozikowski
- Subjects
Indoles ,Mitosis ,Pharmacology ,Histone Deacetylase 6 ,Hydroxamic Acids ,T-Lymphocytes, Regulatory ,Histone Deacetylases ,Mice ,Structure-Activity Relationship ,T-Lymphocyte Subsets ,In vivo ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Cell Proliferation ,Immunosuppression Therapy ,Chemistry ,Effector ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Forkhead Transcription Factors ,HDAC6 ,Recombinant Proteins ,HDAC1 ,In vitro ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Biochemistry ,CD4 Antigens ,Molecular Medicine ,Immunosuppressive Agents ,Carbolines - Abstract
Second-generation Tubastatin A analogues were synthesized and evaluated for their ability to inhibit selectively histone deacetylase 6 (HDAC6). Substitutions to the carboline cap group were well-tolerated with substitution at the 2-position of both β- and γ-carbolines being optimal for HDAC6 activity and selectivity. Some compounds in this series were determined to have subnanomolar activity at HDAC6 with more than 7000 fold selectivity for HDAC6 versus HDAC1. Selected compounds were then evaluated for their ability to augment the immunosuppressive effect of Foxp3+ regulatory T cells. All compounds tested were found to enhance the ability of regulatory T cells to inhibit the mitotic division of effector T cells both in vitro and in vivo, suggesting that further investigation into the use of these compounds for the treatment of autoimmune disorders is warranted.
- Published
- 2012
29. Bicyclic-Capped Histone Deacetylase 6 Inhibitors with Improved Activity in a Model of Axonal Charcot–Marie–Tooth Disease
- Author
-
Alan P. Kozikowski, Giulio Vistoli, Ludo Van Den Bosch, Wanda Haeck, Mariana Frojuello, Veronick Benoy, Sida Shen, Jay H. Kalin, and Joel A. Bergman
- Subjects
0301 basic medicine ,Physiology ,Cognitive Neuroscience ,Mice, Transgenic ,medicine.disease_cause ,Histone Deacetylase 6 ,Biochemistry ,Article ,Histone Deacetylases ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Neuroblastoma ,Charcot-Marie-Tooth Disease ,Tubulin ,Cell Line, Tumor ,Ganglia, Spinal ,medicine ,Animals ,Humans ,Cells, Cultured ,Neurons ,Mutation ,Histone deacetylase 5 ,Hydroxamic acid ,biology ,Acetylation ,Cell Biology ,General Medicine ,HDAC6 ,HDAC1 ,3. Good health ,Cell biology ,Histone Deacetylase Inhibitors ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Histone ,chemistry ,Peripheral nervous system ,biology.protein ,Interleukin-2 - Abstract
Charcot-Marie-Tooth (CMT) disease is a disorder of the peripheral nervous system where progressive degeneration of motor and sensory nerves leads to motor problems and sensory loss and for which no pharmacological treatment is available. Recently, it has been shown in a model for the axonal form of CMT that histone deacetylase 6 (HDAC6) can serve as a target for the development of a pharmacological therapy. Therefore, we aimed at developing new selective and activity-specific HDAC6 inhibitors with improved biochemical properties. By utilizing a bicyclic cap as the structural scaffold from which to build upon, we developed several analogues that showed improved potency compared to tubastatin A while maintaining excellent selectivity compared to HDAC1. Further screening in N2a cells examining both the acetylation of α-tubulin and histones narrowed down the library of compounds to three potent and selective HDAC6 inhibitors. In mutant HSPB1-expressing DRG neurons, serving as an in vitro model for CMT2, these inhibitors were able to restore the mitochondrial axonal transport deficits. Combining structure-based development of HDAC6 inhibitors, screening in N2a cells and in a neuronal model for CMT2F, and preliminary ADMET and pharmacokinetic profiles, resulted in the selection of compound 23d that possesses improved biochemical, functional, and druglike properties compared to tubastatin A.
- Published
- 2015
30. CoREST in Peace: Dual Action Inhibitors of Histone Deacetylase and Lysine Specific Demethylase
- Author
-
Wayne W. Hancock, Justin Roberts, James E. Bradner, Jay H. Kalin, Polina Prusevich, Muzhou Wu, Byungwoo Ryu, Rhoda M. Alani, Liqing Wang, Dawn Hayward, and Philip A. Cole
- Subjects
Histone deacetylase 5 ,biology ,HDAC11 ,Chemistry ,Histone deacetylase 2 ,HDAC10 ,food and beverages ,Biochemistry ,HDAC1 ,Cell biology ,Histone ,Gene expression ,Genetics ,biology.protein ,Histone deacetylase ,Molecular Biology ,Biotechnology - Abstract
Histone deacetylases (HDACs) have been shown to act broadly on thousands of protein lysine acetylation sites to regulate cell growth and gene expression. The class I enzymes HDAC1 and HDAC2 can be ...
- Published
- 2015
31. Abstract 5068: A novel dual action inhibitor of histone deacetylase and demethylase in melanoma
- Author
-
Muzhou Wu, Rhoda M. Alani, Byungwoo Ryu, Jay H. Kalin, and Philip A. Cole
- Subjects
Cancer Research ,Histone deacetylase 5 ,Entinostat ,Histone deacetylase 2 ,HDAC11 ,HDAC10 ,Biology ,medicine.disease_cause ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer research ,biology.protein ,medicine ,Demethylase ,Histone deacetylase ,Carcinogenesis - Abstract
Epigenetic agents have drawn great attention as anti-cancer therapies, with several HDAC inhibitors approved for a subset of hematologic malignancies. One of the biggest challenges in targeting epigenetic mechanisms of tumorigenesis is the wide spectrum of effects which restrict the therapeutic window for these compounds. We have developed a series of potent small molecule inhibitors with specificity towards the CoREST epigenetic corepressor complex through a dual-action mechanism targeting LSD1 and HDAC1. These compounds show a unique profile of pharmacologic action with an improved therapeutic window in a variety of cell types. Screening of tumor cell lines for growth inhibitory effects revealed variable efficacy in a broad spectrum of cancers with the most consistent and potent effects seen in human melanomas. The growth of a number of melanoma cell lines was found to be potently inhibited by one of these compounds, Corin 2; however, primary human melanocytes were relatively resistant to this agent. Transcriptomic analysis revealed that Corin2 was a more potent inducer of tumor suppressor genes compared to the parent HDAC and LSD1 compounds. Genetic knockdown of CoREST or LSD1 in cancer cell lines abolished the differences in potency of Corin2 vs. the parent HDAC inhibitor, Entinostat, suggesting that Corin2's favorable pharmacologic effects rely on an intact CoREST complex. Corin2 was also effective in slowing tumor growth in a melanoma mouse xenograft model. These dual action inhibitors demonstrate a novel, potent, and specific therapeutic approach to targeting epigenetic pathways in human melanomas which may lead to improved therapeutic benefits in patients with advanced disease. Note: This abstract was not presented at the meeting. Citation Format: Muzhou WU, Jay Kalin, Byungwoo Ryu, Philip Cole, Rhoda Alani. A novel dual action inhibitor of histone deacetylase and demethylase in melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5068. doi:10.1158/1538-7445.AM2017-5068
- Published
- 2017
32. A novel role for histone deacetylase 6 in the regulation of the tolerogenic STAT3/IL-10 pathway in APCs
- Author
-
Fengdong Cheng, Calvin Lee, Patricio Perez-Villarroel, Alejandro Villagra, Jay H. Kalin, Jennifer Rock-Klotz, Maritza Lienlaf, Wayne W. Hancock, Jianguo Tao, Edward Seto, Karrune Woan, Eduardo M. Sotomayor, Hongwei Wang, Javier Pinilla-Ibarz, Alan P. Kozikowski, David M. Woods, and Eva Sahakian
- Subjects
CD4-Positive T-Lymphocytes ,Male ,STAT3 Transcription Factor ,Chromatin Immunoprecipitation ,Transcription, Genetic ,T cell ,Immunology ,Antigen-Presenting Cells ,Gene Expression ,Histone Deacetylase 6 ,Lymphocyte Activation ,Histone Deacetylases ,Article ,Immune tolerance ,Cell Line ,Mice ,Immune system ,medicine ,Immune Tolerance ,Immunology and Allergy ,Animals ,Phosphorylation ,RNA, Small Interfering ,Antigen-presenting cell ,Promoter Regions, Genetic ,Regulation of gene expression ,Inflammation ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Macrophages ,Acetylation ,Dendritic Cells ,Molecular biology ,Recombinant Proteins ,Cell biology ,Interleukin-10 ,Protein Structure, Tertiary ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Interleukin 10 ,Histone ,medicine.anatomical_structure ,Gene Expression Regulation ,Mutation ,biology.protein ,RNA Interference ,Chromatin immunoprecipitation ,Oligopeptides - Abstract
APCs are critical in T cell activation and in the induction of T cell tolerance. Epigenetic modifications of specific genes in the APC play a key role in this process, and among them histone deacetylases (HDACs) have emerged as key participants. HDAC6, one of the members of this family of enzymes, has been shown to be involved in regulation of inflammatory and immune responses. In this study, to our knowledge we show for the first time that genetic or pharmacologic disruption of HDAC6 in macrophages and dendritic cells results in diminished production of the immunosuppressive cytokine IL-10 and induction of inflammatory APCs that effectively activate Ag-specific naive T cells and restore the responsiveness of anergic CD4+ T cells. Mechanistically, we have found that HDAC6 forms a previously unknown molecular complex with STAT3, association that was detected in both the cytoplasmic and nuclear compartments of the APC. By using HDAC6 recombinant mutants we identified the domain comprising amino acids 503–840 as being required for HDAC6 interaction with STAT3. Furthermore, by re–chromatin immunoprecipitation we confirmed that HDAC6 and STAT3 are both recruited to the same DNA sequence within the Il10 gene promoter. Of note, disruption of this complex by knocking down HDAC6 resulted in decreased STAT3 phosphorylation—but no changes in STAT3 acetylation—as well as diminished recruitment of STAT3 to the Il10 gene promoter region. The additional demonstration that a selective HDAC6 inhibitor disrupts this STAT3/IL-10 tolerogenic axis points to HDAC6 as a novel molecular target in APCs to overcome immune tolerance and tips the balance toward T cell immunity.
- Published
- 2014
33. RI-1: a chemical inhibitor of RAD51 that disrupts homologous recombination in human cells
- Author
-
Douglas K. Bishop, Anna Zelivianskaia, Hillary L. Logan, Jay H. Kalin, Jeremy M. Stark, Luke L. Miller, Alan P. Kozikowski, Philip P. Connell, Brian Budke, and William Cameron McGuire
- Subjects
DNA repair ,DNA damage ,Mitomycin ,Morpholines ,RAD51 ,Biology ,Genome Integrity, Repair and Replication ,Homology directed repair ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,Humans ,Pyrroles ,Cysteine ,Enzyme Inhibitors ,Homologous Recombination ,Replication protein A ,030304 developmental biology ,0303 health sciences ,Antibiotics, Antineoplastic ,DNA repair protein XRCC4 ,Molecular biology ,Cell biology ,Protein Subunits ,Cross-Linking Reagents ,chemistry ,030220 oncology & carcinogenesis ,Rad51 Recombinase ,Homologous recombination ,DNA ,DNA Damage - Abstract
Homologous recombination serves multiple roles in DNA repair that are essential for maintaining genomic stability. We here describe RI-1, a small molecule that inhibits the central recombination protein RAD51. RI-1 specifically reduces gene conversion in human cells while stimulating single strand annealing. RI-1 binds covalently to the surface of RAD51 protein at cysteine 319 that likely destabilizes an interface used by RAD51 monomers to oligomerize into filaments on DNA. Correspondingly, the molecule inhibits the formation of subnuclear RAD51 foci in cells following DNA damage, while leaving replication protein A focus formation unaffected. Finally, it potentiates the lethal effects of a DNA cross-linking drug in human cells. Given that this inhibitory activity is seen in multiple human tumor cell lines, RI-1 holds promise as an oncologic drug. Furthermore, RI-1 represents a unique tool to dissect the network of reaction pathways that contribute to DNA repair in cells.
- Published
- 2012
34. Chiral mercaptoacetamides display enantioselective inhibition of histone deacetylase 6 and exhibit neuroprotection in cortical neuron models of oxidative stress
- Author
-
Jay H. Kalin, Sophie Gaudrel-Grosay, Alan P. Kozikowski, Giulio Vistoli, and Hankun Zhang
- Subjects
Gene isoform ,Models, Molecular ,Stereochemistry ,Primary Cell Culture ,medicine.disease_cause ,Histone Deacetylase 6 ,Hydroxamic Acids ,Biochemistry ,Neuroprotection ,Histone Deacetylases ,Stereocenter ,chemistry.chemical_compound ,Drug Discovery ,Acetamides ,medicine ,Animals ,Humans ,Protein Isoforms ,Sulfhydryl Compounds ,General Pharmacology, Toxicology and Pharmaceutics ,Pharmacology ,Neurons ,Hydroxamic acid ,Chemistry ,Organic Chemistry ,Enantioselective synthesis ,Neurodegenerative Diseases ,Stereoisomerism ,HDAC6 ,Rats ,Histone Deacetylase Inhibitors ,Oxidative Stress ,Neuroprotective Agents ,Molecular Medicine ,Histone deacetylase ,Dimerization ,Oxidative stress - Abstract
Mercaptoacetamide-based ligands have been designed as a new class of histone deacetylase (HDAC) inhibitors for possible use in the treatment of neurodegenerative diseases. The thiol group of these compounds provides a key binding element for interaction with the catalytic zinc ion, and thus differs from the more typically employed hydroxamic acid based zinc binding groups. Herein we disclose the chemistry and biology of some substituted mercaptoacetamides with the intention of increasing HDAC6 isoform selectivity while maintaining potency similar to their hydroxamic acid analogues. The introduction of a stereocenter α to the thiol group was found to have a considerable impact on HDAC inhibitor potency. These new compounds were also profiled for their therapeutic potential in an in vitro model of stress-induced neuronal injury and were found to act as nontoxic neuroprotective agents.
- Published
- 2011
35. Rational Design and Simple Chemistry Yield a Superior, Neuroprotective HDAC6 Inhibitor, Tubastatin A
- Author
-
Jay H. Kalin, Camille Brochier, Brett Langley, Kyle V. Butler, Guilio Vistoli, and Alan P. Kozikowski
- Subjects
Models, Molecular ,Indoles ,medicine.disease_cause ,Histone Deacetylase 6 ,Hydroxamic Acids ,Biochemistry ,Neuroprotection ,Catalysis ,Article ,Histone Deacetylases ,chemistry.chemical_compound ,Structure-Activity Relationship ,Colloid and Surface Chemistry ,In vivo ,medicine ,Structure–activity relationship ,Humans ,Anilides ,Enzyme Inhibitors ,Homocysteine ,Neurons ,Cell Death ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Rational design ,Stereoisomerism ,General Chemistry ,Glutathione ,HDAC6 ,Isoenzymes ,Oxidative Stress ,Neuroprotective Agents ,Acetylation ,Drug Design ,Oxidative stress - Abstract
Structure-based drug design combined with homology modeling techniques were used to develop potent inhibitors of HDAC6 that display superior selectivity for the HDAC6 isozyme compared to other inhibitors. These inhibitors can be assembled in a few synthetic steps, and thus are readily scaled up for in vivo studies. An optimized compound from this series, designated Tubastatin A, was tested in primary cortical neuron cultures in which it was found to induce elevated levels of acetylated alpha-tubulin, but not histone, consistent with its HDAC6 selectivity. Tubastatin A also conferred dose-dependent protection in primary cortical neuron cultures against glutathione depletion-induced oxidative stress. Importantly, when given alone at all concentrations tested, this hydroxamate-containing HDAC6-selective compound displayed no neuronal toxicity, thus, forecasting the potential application of this agent and its analogues to neurodegenerative conditions.
- Published
- 2010
36. Creating zinc monkey wrenches in the treatment of epigenetic disorders
- Author
-
Jay H. Kalin, Kyle V. Butler, and Alan P. Kozikowski
- Subjects
Rett syndrome ,Disease ,Pharmacology ,Bioinformatics ,Hydroxamic Acids ,Biochemistry ,Models, Biological ,Histone Deacetylases ,Analytical Chemistry ,Epigenesis, Genetic ,Prostate cancer ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Bipolar disorder ,Enzyme Inhibitors ,Histone Acetyltransferases ,business.industry ,Mood Disorders ,Multiple sclerosis ,Neurodegenerative Diseases ,medicine.disease ,Histone Deacetylase Inhibitors ,Zinc ,Mood disorders ,Schizophrenia ,Drug Therapy, Combination ,Histone deacetylase ,business - Abstract
The approval of suberoylanilide hydroxamic acid by the FDA for the treatment of cutaneous T-cell lymphoma in October, 2006 sparked a dramatic increase in the development of inhibitors for the class of enzymes known as the histone deacetylases (HDACs). In recent years, a large number of combination therapies involving histone deacetylase inhibitors (HDACIs) have been developed for the treatment of a variety of malignancies and neurodegenerative disorders. Promising evidence has been reported for the treatment of pancreatic cancer, prostate cancer, and leukemia as well as a number of other previously difficult to treat cancers. Drug combination approaches have also shown promise for the treatment of mood disorders including bipolar disorder and depression. In addition to these drug combination approaches, HDACIs alone have demonstrated effectiveness in the treatment of Parkinson's disease, Alzheimer's disease, Rubinstein–Taybi syndrome, Rett syndrome, Friedreich's ataxia, Huntington's disease, multiple sclerosis, anxiety, and schizophrenia. Adverse inflammatory affects observed with traumatic brain injury and arthritis have also been alleviated by treatment with certain HDACIs. Based on the diverse utility and wide range of mechanistic actions observed with this class of drugs, the future development of better drug combination therapies and more selective HDACIs is warranted.
- Published
- 2009
37. Synthesis and antitubercular activity of quaternized promazine and promethazine derivatives
- Author
-
Erica L. Amorose, Yuehong Wang, Sang-Hyun Cho, Eric M. Fooksman, Michael John Rodig, Miguel O. Mitchell, Heather M. Williams, Aaron B. Bate, Cristofer M. Price, Scott G. Franzblau, and Jay H. Kalin
- Subjects
Stereochemistry ,Chlorpromazine ,Clinical Biochemistry ,Substituent ,Antitubercular Agents ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Promethazine ,chemistry.chemical_compound ,Structure-Activity Relationship ,Bromide ,Phenothiazine ,Drug Discovery ,medicine ,Molecular Biology ,Promazine ,Antibacterial agent ,Triflupromazine ,Organic Chemistry ,Mycobacterium tuberculosis ,Quaternary Ammonium Compounds ,chemistry ,Molecular Medicine ,medicine.drug - Abstract
Quaternized chlorpromazine, triflupromazine, and promethazine derivatives were synthesized and examined as antitubercular agents against both actively growing and non-replicating Mycobacterium tuberculosis H37Rv. Impressively, several compounds inhibited non-replicating M. tuberculosis at concentrations equal to or double their MICs against the actively growing strain. All active compounds were non-toxic toward Vero cells (IC 50 > 128 μM). N -Allylchlorpromazinium bromide was only weakly antitubercular, but replacing allyl with benzyl or substituted benzyl improved potency. An electron-withdrawing substituent on the phenothiazine ring was also essential. Branching at the carbon chain decreased antitubercular activity. The optimum antitubercular structures possessed N -(4- or 3-chlorobenzyl) substitution on triflupromazine.
- Published
- 2006
38. Histone deacetylase 6 inhibition improves memory and reduces total tau levels in a mouse model of tau deposition
- Author
-
Maj Linda B. Selenica, Alan P. Kozikowski, Joel A. Bergman, Jay H. Kalin, Steven B. Housley, Dave Morgan, Leif Benner, Marcia N. Gordon, Daniel C. Lee, Kevin Nash, and Barbara Manchec
- Subjects
Pathology ,medicine.medical_specialty ,biology ,business.industry ,Research ,Cognitive Neuroscience ,Neurodegeneration ,Clinical Neurology ,HDAC6 ,medicine.disease ,Hsp90 ,Cell biology ,Histone ,Neurology ,Acetylation ,Memory improvement ,Heat shock protein ,medicine ,biology.protein ,Neurology (clinical) ,Tauopathy ,business - Abstract
Introduction Tau pathology is associated with a number of age-related neurodegenerative disorders. Few treatments have been demonstrated to diminish the impact of tau pathology in mouse models and none are yet effective in humans. Histone deacetylase 6 (HDAC6) is an enzyme that removes acetyl groups from cytoplasmic proteins, rather than nuclear histones. Its substrates include tubulin, heat shock protein 90 and cortactin. Tubastatin A is a selective inhibitor of HDAC6. Modification of tau pathology by specific inhibition of HDAC6 presents a potential therapeutic approach in tauopathy. Methods We treated rTg4510 mouse models of tau deposition and non-transgenic mice with tubastatin (25 mg/kg) or saline (0.9%) from 5 to 7 months of age. Cognitive behavior analysis, histology and biochemical analysis were applied to access the effect of tubastatin on memory, tau pathology and neurodegeneration (hippocampal volume). Results We present data showing that tubastatin restored memory function in rTg4510 mice and reversed a hyperactivity phenotype. We further found that tubastatin reduced the levels of total tau, both histologically and by western analysis. Reduction in total tau levels was positively correlated with memory improvement in these mice. However, there was no impact on phosphorylated forms of tau, either by histology or western analysis, nor was there an impact on silver positive inclusions histologically. Conclusion Potential mechanisms by which HDAC6 inhibitors might benefit the rTg4510 mouse include stabilization of microtubules secondary to increased tubulin acetylation, increased degradation of tau secondary to increased acetylation of HSP90 or both. These data support the use of HDAC6 inhibitors as potential therapeutic agents against tau pathology.
- Published
- 2014
39. Abstract 3555: Histone deacetylase 6 as a novel regulator of the immunogenicity and aggressiveness of melanoma
- Author
-
Eduardo M. Sotomayor, Zi Wang, Eva Sahakian, Joel A. Bergman, Danay Marante, Alejandro Villagra, Fengdong Cheng, Jay H. Kalin, Karrune Woan, Ajaypal S. Gill, Javier Pinilla-Ibarz, Edward Seto, John Powers, Maritza Lienlaf, Jennifer Rock-Klotz, Alan P. Kozikowski, Jane L. Messina, Hongwei Wang, Alex Achille, Patricio Perez, David M. Woods, Lianet Vazquez, and Jeffrey S. Weber
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Melanoma ,Cancer ,Ipilimumab ,Immunotherapy ,Acquired immune system ,medicine.disease ,Immune tolerance ,Immune system ,Oncology ,MHC class I ,medicine ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Melanoma is currently the fastest growing cancer in incidence according to the World Health Organization. Currently, few therapies provide significant prolongation of survival for metastatic melanoma. Immunotherapy is an attractive modality with potentially few side effects due to the antigen specificity of adaptive immunity. The latest therapy approved by the FDA for the treatment of melanoma was ipilimumab, an antibody against CTLA-4, a key regulator of T-cell activity; however, this therapy offers modest improvements in overall survival. Overcoming mechanisms of tumor-mediated immune suppression requires targeting multiple pathways. One strategy that has gained attention has been the use of histone deacetylase inhibitors (HDACi). Indeed, HDACi treatment has been shown to augment the expression of immunologically relevant genes such as MHC and costimulatory molecules. Furthermore, we have previously demonstrated inhibition of IL-10, a potent anti-inflammatory cytokine, upon treatment of macrophages with HDACi. However, most studies to date have used pan-HDACi, which inhibit all 11 zinc-dependent HDACs. Therefore, the use of more selective HDACi might be preferable in order to minimize side effects. Herein, we demonstrate that HDAC6 is a molecular target in melanoma. Both pharmacologic and genetic disruption of HDAC6 in B16 murine melanoma cells using HDAC6-selective inhibitors (HDAC6i) and targeted shRNA (HDAC6KD), respectively, led to inhibition of proliferation, characterized by G1 arrest measured by propidium iodine staining for DNA content. Furthermore, treatment with HDAC6i led to enhanced expression of immunologically relevant receptors including MHC I and MHC II. In vivo, subcutaneous injection in wild type mice of HDAC6KD B16 cells led to delayed tumor growth as compared with control cells. However, this effect was abrogated in experiments using SCID mice, which lack T- and B-cells, suggesting a critical immune component for tumor control in vivo. The mechanism(s) by which HDAC6 regulates tumor immunogenicity are yet to be defined. One possible mechanism arises from protein immunoprecipitation studies which demonstrate that HDAC6 interacts with, and potentially regulates of STAT3, an important survival and pathogenic factor in melanoma, which also has implications for immune tolerance. Finally, the expression HDAC6 was found to be upregulated in a majority of melanoma patient tumor biopsies by gene microarray analysis, as compared with normal skin. This observation was supported by immunohistochemically-stained patient melanoma tissue microarray. Taken together, HDAC6 presents as an attractive therapeutic target in melanoma by both delaying tumor growth and conferring a more attractive immune target, providing rationale for the development and use of selective HDAC6i. Moreover, the current availability of HDAC6i enhances the translational implications of this work. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3555. doi:1538-7445.AM2012-3555
- Published
- 2012
40. Development and characterization of rabbit monoclonal antibodies that recognize human spermine oxidase and application to immunohistochemistry of human cancer tissues.
- Author
-
Armand W J W Tepper, Gerald Chu, Vincent N A Klaren, Jay H Kalin, Patricia Molina-Ortiz, and Antonietta Impagliazzo
- Subjects
Medicine ,Science - Abstract
The enzyme spermine oxidase (SMOX) is involved in polyamine catabolism and converts spermine to spermidine. The enzymatic reaction generates reactive hydrogen peroxide and aldehydes as by-products that can damage DNA and other biomolecules. Increased expression of SMOX is frequently found in lung, prostate, colon, stomach and liver cancer models, and the enzyme also appears to play a role in neuronal dysfunction and vascular retinopathy. Because of growing evidence that links SMOX activity with DNA damage, inflammation, and carcinogenesis, the enzyme has come into view as a potential drug target. A major challenge in cancer research is the lack of characterization of antibodies used for identification of target proteins. To overcome this limitation, we generated a panel of high-affinity rabbit monoclonal antibodies against various SMOX epitopes and selected antibodies for use in immunoblotting, SMOX quantification assays, immunofluorescence microscopy and immunohistochemistry. Immunohistochemistry analysis with the antibody SMAB10 in normal and transformed tissues confirms that SMOX is upregulated in several different cancers. Together, the panel of antibodies generated herein adds to the toolbox of high-quality reagents to study SMOX biology and to facilitate SMOX drug development.
- Published
- 2022
- Full Text
- View/download PDF
41. Investigation into the use of histone deacetylase inhibitor MS-275 as a topical agent for the prevention and treatment of cutaneous squamous cell carcinoma in an SKH-1 hairless mouse model.
- Author
-
Jay H Kalin, Abdulkerim Eroglu, Hua Liu, W David Holtzclaw, Irene Leigh, Charlotte M Proby, Jed W Fahey, Philip A Cole, and Albena T Dinkova-Kostova
- Subjects
Medicine ,Science - Abstract
Cutaneous squamous cell carcinomas are a common form of highly mutated keratinocyte skin cancers that are of particular concern in immunocompromised patients. Here we report on the efficacy of topically applied MS-275, a clinically used histone deacetylase inhibitor, for the treatment and management of this disease. At 2 mg/kg, MS-275 significantly decreased tumor burden in an SKH-1 hairless mouse model of UVB radiation-induced skin carcinogenesis. MS-275 was cell permeable as a topical formulation and induced histone acetylation changes in mouse tumor tissue. MS-275 was also effective at inhibiting the proliferation of patient derived cutaneous squamous cell carcinoma lines and was particularly potent toward cells isolated from a regional metastasis on an immunocompromised individual. Our findings support the use of alternative routes of administration for histone deacetylase inhibitors in the treatment of high-risk squamous cell carcinoma which may ultimately lead to more precise delivery and reduced systemic toxicity.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.