303 results on '"Andrei V. Gudkov"'
Search Results
2. Signaling through TLR5 mitigates lethal radiation damage by neutrophil-dependent release of MMP-9
- Author
-
Craig M. Brackett, Kellee F. Greene, Alyssa R. Aldrich, Nicholas H. Trageser, Srabani Pal, Ivan Molodtsov, Bojidar M. Kandar, Lyudmila G. Burdelya, Scott I. Abrams, and Andrei V. Gudkov
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Acute radiation syndrome (ARS) is a major cause of lethality following radiation disasters. A TLR5 agonist, entolimod, is among the most powerful experimental radiation countermeasures and shows efficacy in rodents and non-human primates as a prophylactic (radioprotection) and treatment (radiomitigation) modality. While the prophylactic activity of entolimod has been connected to the suppression of radiation-induced apoptosis, the mechanism by which entolimod functions as a radiomitigator remains poorly understood. Uncovering this mechanism has significant and broad-reaching implications for the clinical development and improvement of TLR5 agonists for use as an effective radiation countermeasure in scenarios of mass casualty resulting from accidental exposure to ionizing radiation. Here, we demonstrate that in contrast to radioprotection, neutrophils are essential for the radiomitigative activity of entolimod in a mouse model of lethal ARS. Neutrophils express functional TLR5 and rapidly exit the bone marrow (BM), accumulate in solid tissues, and release MMP-9 following TLR5 stimulation which is accompanied by an increase in the number of active hematopoietic pluripotent precursors (HPPs) in the BM. Importantly, recombinant MMP-9 by itself has radiomitigative activity and, in the absence of neutrophils, accelerates the recovery of the hematopoietic system. Unveiling this novel TLR5-neutrophil-MMP-9 axis of radiomitigation opens new opportunities for the development of efficacious radiation countermeasures to treat ARS following accidental radiation disasters.
- Published
- 2021
- Full Text
- View/download PDF
3. Resistance of bone marrow stroma to genotoxic preconditioning is determined by p53
- Author
-
Natalia Fedtsova, Elena A. Komarova, Kellee F. Greene, Liliya R. Novototskaya, Ivan Molodtsov, Craig M. Brackett, Evguenia Strom, Anatoli S. Gleiberman, Alexander N. Shakhov, and Andrei V. Gudkov
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Transplantation of bone marrow (BM) is made possible by the differential sensitivity of its stromal and hematopoietic components to preconditioning by radiation and/or chemotherapeutic drugs. These genotoxic treatments eliminate host hematopoietic precursors by inducing p53-mediated apoptosis but keep the stromal niche sufficiently intact for the engraftment of donor hematopoietic cells. We found that p53-null mice cannot be rescued by BM transplantation (BMT) from even the lowest lethal dose of total body irradiation (TBI). We compared structural changes in BM stroma of mice differing in their p53 status to understand why donor BM failed to engraft in the irradiated p53-null mice. Irradiation did not affect the general structural integrity of BM stroma and induced massive expression of alpha-smooth muscle actin in mesenchymal cells followed by increased adiposity in p53 wild-type mice. In contrast, none of these events were found in p53-null mice, whose BM stroma underwent global structural damage following TBI. Similar differences in response to radiation were observed in in vitro-grown bone-adherent mesenchymal cells (BAMC): p53-null cells underwent mitotic catastrophe while p53 wild-type cells stayed arrested but viable. Supplementation with intact BAMC of either genotype enabled donor BM engraftment and significantly extended longevity of irradiated p53-null mice. Thus, successful preconditioning depends on the p53-mediated protection of cells critical for the functionality of BM stroma. Overall, this study reveals a dual positive role of p53 in BMT: it drives apoptotic death of hematopoietic cells and protects BM stromal cells essential for its functionality.
- Published
- 2021
- Full Text
- View/download PDF
4. Longitudinal analysis of blood markers reveals progressive loss of resilience and predicts human lifespan limit
- Author
-
Timothy V. Pyrkov, Konstantin Avchaciov, Andrei E. Tarkhov, Leonid I. Menshikov, Andrei V. Gudkov, and Peter O. Fedichev
- Subjects
Science - Abstract
Aging is associated with an increased risk of chronic diseases and functional decline. Here, the authors investigate the fluctuations of physiological indices along aging trajectories and observed a characteristic decrease in the organism state recovery rate.
- Published
- 2021
- Full Text
- View/download PDF
5. A deimmunized and pharmacologically optimized Toll-like receptor 5 agonist for therapeutic applications
- Author
-
Vadim Mett, Oleg V. Kurnasov, Ivan A. Bespalov, Ivan Molodtsov, Craig M. Brackett, Lyudmila G. Burdelya, Andrei A. Purmal, Anatoli S. Gleiberman, Ilia A. Toshkov, Catherine A. Burkhart, Yakov N. Kogan, Ekaterina L. Andrianova, Andrei V. Gudkov, and Andrei L. Osterman
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Mett et al. describe development of GP532, a substantially deimmunized derivative of Toll-like receptor 5 (TLR5) agonist entolimod. GP532 has mutations eliminating key B- and T-cell epitopes and an inflammasome-activating domain yet remains a potent NF-κB activator with biological effects similar to entolimod. Thus, GP532 is suitable for multi-dose TLR5-targeting therapies and patients with high titers of preexisting flagellin-neutralizing antibodies.
- Published
- 2021
- Full Text
- View/download PDF
6. The Combination of Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Delays KMT2A-Rearranged Leukemia Progression
- Author
-
Lin Xiao, Mawar Karsa, Emma Ronca, Angelika Bongers, Angelika Kosciolek, Ali El-Ayoubi, Jezrael L. Revalde, Janith A. Seneviratne, Belamy B. Cheung, Laurence C. Cheung, Rishi S. Kotecha, Andrea Newbold, Stefan Bjelosevic, Greg M. Arndt, Richard B. Lock, Ricky W. Johnstone, Andrei V. Gudkov, Katerina V. Gurova, Michelle Haber, Murray D. Norris, Michelle J. Henderson, and Klaartje Somers
- Subjects
KMT2A-rearranged leukemia ,infant leukemia ,curaxin CBL0137 ,histone deacetylase inhibition ,chromatin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Rearrangements of the Mixed Lineage Leukemia (MLL/KMT2A) gene are present in approximately 10% of acute leukemias and characteristically define disease with poor outcome. Driven by the unmet need to develop better therapies for KMT2A-rearranged leukemia, we previously discovered that the novel anti-cancer agent, curaxin CBL0137, induces decondensation of chromatin in cancer cells, delays leukemia progression and potentiates standard of care chemotherapies in preclinical KMT2A-rearranged leukemia models. Based on the promising potential of histone deacetylase (HDAC) inhibitors as targeted anti-cancer agents for KMT2A-rearranged leukemia and the fact that HDAC inhibitors also decondense chromatin via an alternate mechanism, we investigated whether CBL0137 could potentiate the efficacy of the HDAC inhibitor panobinostat in KMT2A-rearranged leukemia models. The combination of CBL0137 and panobinostat rapidly killed KMT2A-rearranged leukemia cells by apoptosis and significantly delayed leukemia progression and extended survival in an aggressive model of MLL-AF9 (KMT2A:MLLT3) driven murine acute myeloid leukemia. The drug combination also exerted a strong anti-leukemia response in a rapidly progressing xenograft model derived from an infant with KMT2A-rearranged acute lymphoblastic leukemia, significantly extending survival compared to either monotherapy. The therapeutic enhancement between CBL0137 and panobinostat in KMT2A-r leukemia cells does not appear to be mediated through cooperative effects of the drugs on KMT2A rearrangement-associated histone modifications. Our data has identified the CBL0137/panobinostat combination as a potential novel targeted therapeutic approach to improve outcome for KMT2A-rearranged leukemia.
- Published
- 2022
- Full Text
- View/download PDF
7. Dual targeting of the epigenome via FACT complex and histone deacetylase is a potent treatment strategy for DIPG
- Author
-
Anahid Ehteda, Sandy Simon, Laura Franshaw, Federico M. Giorgi, Jie Liu, Swapna Joshi, Jourdin R.C. Rouaen, Chi Nam Ignatius Pang, Ruby Pandher, Chelsea Mayoh, Yujie Tang, Aaminah Khan, Caitlin Ung, Ornella Tolhurst, Anne Kankean, Elisha Hayden, Rebecca Lehmann, Sylvie Shen, Anjana Gopalakrishnan, Peter Trebilcock, Katerina Gurova, Andrei V. Gudkov, Murray D. Norris, Michelle Haber, Orazio Vittorio, Maria Tsoli, and David S. Ziegler
- Subjects
DIPG ,brainstem glioma ,H3K27M ,facilitates chromatin transcription complex ,HDAC ,EZH2 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Diffuse intrinsic pontine glioma (DIPG) is an aggressive and incurable childhood brain tumor for which new treatments are needed. CBL0137 is an anti-cancer compound developed from quinacrine that targets facilitates chromatin transcription (FACT), a chromatin remodeling complex involved in transcription, replication, and DNA repair. We show that CBL0137 displays profound cytotoxic activity against a panel of patient-derived DIPG cultures by restoring tumor suppressor TP53 and Rb activity. Moreover, in an orthotopic model of DIPG, treatment with CBL0137 significantly extends animal survival. The FACT subunit SPT16 is found to directly interact with H3.3K27M, and treatment with CBL0137 restores both histone H3 acetylation and trimethylation. Combined treatment of CBL0137 with the histone deacetylase inhibitor panobinostat leads to inhibition of the Rb/E2F1 pathway and induction of apoptosis. The combination of CBL0137 and panobinostat significantly prolongs the survival of mice bearing DIPG orthografts, suggesting a potential treatment strategy for DIPG.
- Published
- 2021
- Full Text
- View/download PDF
8. A Purine Nucleotide Biosynthesis Enzyme Guanosine Monophosphate Reductase Is a Suppressor of Melanoma Invasion
- Author
-
Joseph A. Wawrzyniak, Anna Bianchi-Smiraglia, Wiam Bshara, Sudha Mannava, Jeff Ackroyd, Archis Bagati, Angela R. Omilian, Michael Im, Natalia Fedtsova, Jeffrey C. Miecznikowski, Kalyana C. Moparthy, Shoshanna N. Zucker, Qianqian Zhu, Nadezhda I. Kozlova, Albert E. Berman, Keith S. Hoek, Andrei V. Gudkov, Donna S. Shewach, Carl D. Morrison, and Mikhail A. Nikiforov
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.
- Published
- 2013
- Full Text
- View/download PDF
9. Supplementary Materials from Targeting FACT Complex Suppresses Mammary Tumorigenesis in Her2/neu Transgenic Mice
- Author
-
Katerina V. Gurova, Andrei V. Gudkov, Carl Morrison, Song Liu, Dan Wang, Andrei A. Purmal, Ilya Toshkov, Alfiya Safina, Srabani Pal, Bhupinder Hoonjan, Geraldine Paszkiewicz, Mairead Commane, and Igor E. Koman
- Abstract
PDF file - 11K, List of antibodies used in the study
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 1 from Targeting FACT Complex Suppresses Mammary Tumorigenesis in Her2/neu Transgenic Mice
- Author
-
Katerina V. Gurova, Andrei V. Gudkov, Carl Morrison, Song Liu, Dan Wang, Andrei A. Purmal, Ilya Toshkov, Alfiya Safina, Srabani Pal, Bhupinder Hoonjan, Geraldine Paszkiewicz, Mairead Commane, and Igor E. Koman
- Abstract
PDF file - 790K, Absence of morphological changes in organs of MMTV-neu mice treated with Curaxin-137 from 4 to 14 weeks of age.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Prevention of Colorectal Carcinogenesis by DNA-Binding Small-Molecule Curaxin CBL0137 Involves Suppression of Wnt Signaling
- Author
-
Marianna G. Yakubovskaya, Katerina V. Gurova, Andrei V. Gudkov, Gennady A. Belitsky, Andrei A. Purmal, Andrey Leonov, Roman Ivanov, Elena Shipaeva, Ramiz Salimov, Daria Fleyshman, Alfiya Safina, Olga Morozova, Tatiana Gor'kova, Elena Antoshina, Lubov Trukhanova, Varvara Maksimova, Ekaterina A. Lesovaya, Timur Fetisov, and Kirill Kirsanov
- Abstract
Chemoprevention is considered a valid approach to reduce the incidence of colorectal cancer, one of the most common malignancies worldwide. Here, we investigated the tumor-preventive activity of curaxin CBL0137. This compound represents a new class of nonmutagenic DNA-binding small molecules that alter chromatin stability and inhibit the function of the histone chaperone FACT. Among downstream effects of CBL0137 treatment are activation of p53 and type I interferons and inhibition of NFκB, HSF1, and MYC. In addition, our data show that in both human and mouse colorectal cancer cells in vitro, CBL0137 inhibits the APC/WNT/β-catenin signaling pathway, which plays a key role in colon carcinogenesis. Using quantitative RT-PCR and microarray hybridization, we have demonstrated decreased expression of multiple components and downstream targets of the WNT pathway in colon cancer cells treated with CBL0137. At the same time, CBL0137 induced expression of WNT antagonists. Inhibition of WNT signaling activity by CBL0137 was also confirmed by luciferase reporter assay. Tumor-preventive activity of CBL0137 in vivo was tested in a murine model of colorectal carcinogenesis induced by 1,2-dimethylhydrazine (DMH), which is known to involve WNT pathway dysregulation. After DMH subcutaneous treatment, mice were administered CBL0137 in drinking water. Efficacy of CBL0137 in suppressing development of colorectal cancer in this model was evidenced by reduced incidence of adenocarcinomas and adenomas in both males and females and decrease in tumor multiplicity. These data support the prospective use of CBL0137 in chemoprevention of colorectal cancer as well as of other malignances associated with activated WNT signaling.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Tables 1-2 from Targeting FACT Complex Suppresses Mammary Tumorigenesis in Her2/neu Transgenic Mice
- Author
-
Katerina V. Gurova, Andrei V. Gudkov, Carl Morrison, Song Liu, Dan Wang, Andrei A. Purmal, Ilya Toshkov, Alfiya Safina, Srabani Pal, Bhupinder Hoonjan, Geraldine Paszkiewicz, Mairead Commane, and Igor E. Koman
- Abstract
PDF file - 175K, Table 1. Concentration of Curaxin-137 (muM) in mouse plasma and tissues (mean +/-SD in muM, n=4) 10 weeks after start of treatment (14 weeks of age) Table 2. Histopathological findings in old animals in each of treatment group (two mice per group)
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table 5 from Prevention of Colorectal Carcinogenesis by DNA-Binding Small-Molecule Curaxin CBL0137 Involves Suppression of Wnt Signaling
- Author
-
Marianna G. Yakubovskaya, Katerina V. Gurova, Andrei V. Gudkov, Gennady A. Belitsky, Andrei A. Purmal, Andrey Leonov, Roman Ivanov, Elena Shipaeva, Ramiz Salimov, Daria Fleyshman, Alfiya Safina, Olga Morozova, Tatiana Gor'kova, Elena Antoshina, Lubov Trukhanova, Varvara Maksimova, Ekaterina A. Lesovaya, Timur Fetisov, and Kirill Kirsanov
- Abstract
Supplemental Table 5
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Video S2 from Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Michelle Haber, Murray D. Norris, Katerina V. Gurova, Andrei V. Gudkov, David S. Ziegler, Anthony J. Cesare, Ricky W. Johnstone, Glenn Marshall, Jamie I. Fletcher, Michelle J. Henderson, Daniel R. Carter, Andrew J. Gifford, Alvin Kamili, Lei Zhai, Sophie Allan, Stephanie Alfred, Georgina Eden, Adam Kearns, Erin Mosmann, Jessica A. Pettitt, Frances K. Kusuma, Jennifer Brand, Hazel Quek, Pooja Venkat, Chelsea Mayoh, Aisling O'Connor, Katerina I. Leonova, Natalia Issaeva, Laura D. Gamble, Anahid Ehteda, Rachael Terry, Angelika Bongers, Emma Ronca, Mawar Karsa, Ruby Pandher, Jayne Murray, Klaartje Somers, and Lin Xiao
- Abstract
HT10806TG_CBL0137 treatment
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure Legends from Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Michelle Haber, Murray D. Norris, Katerina V. Gurova, Andrei V. Gudkov, David S. Ziegler, Anthony J. Cesare, Ricky W. Johnstone, Glenn Marshall, Jamie I. Fletcher, Michelle J. Henderson, Daniel R. Carter, Andrew J. Gifford, Alvin Kamili, Lei Zhai, Sophie Allan, Stephanie Alfred, Georgina Eden, Adam Kearns, Erin Mosmann, Jessica A. Pettitt, Frances K. Kusuma, Jennifer Brand, Hazel Quek, Pooja Venkat, Chelsea Mayoh, Aisling O'Connor, Katerina I. Leonova, Natalia Issaeva, Laura D. Gamble, Anahid Ehteda, Rachael Terry, Angelika Bongers, Emma Ronca, Mawar Karsa, Ruby Pandher, Jayne Murray, Klaartje Somers, and Lin Xiao
- Abstract
Supplementary Figure Legends S1-S16
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Michelle Haber, Murray D. Norris, Katerina V. Gurova, Andrei V. Gudkov, David S. Ziegler, Anthony J. Cesare, Ricky W. Johnstone, Glenn Marshall, Jamie I. Fletcher, Michelle J. Henderson, Daniel R. Carter, Andrew J. Gifford, Alvin Kamili, Lei Zhai, Sophie Allan, Stephanie Alfred, Georgina Eden, Adam Kearns, Erin Mosmann, Jessica A. Pettitt, Frances K. Kusuma, Jennifer Brand, Hazel Quek, Pooja Venkat, Chelsea Mayoh, Aisling O'Connor, Katerina I. Leonova, Natalia Issaeva, Laura D. Gamble, Anahid Ehteda, Rachael Terry, Angelika Bongers, Emma Ronca, Mawar Karsa, Ruby Pandher, Jayne Murray, Klaartje Somers, and Lin Xiao
- Abstract
Purpose:We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma.Experimental Design:The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction.Results:The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination.Conclusions:The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figures from Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Michelle Haber, Murray D. Norris, Katerina V. Gurova, Andrei V. Gudkov, David S. Ziegler, Anthony J. Cesare, Ricky W. Johnstone, Glenn Marshall, Jamie I. Fletcher, Michelle J. Henderson, Daniel R. Carter, Andrew J. Gifford, Alvin Kamili, Lei Zhai, Sophie Allan, Stephanie Alfred, Georgina Eden, Adam Kearns, Erin Mosmann, Jessica A. Pettitt, Frances K. Kusuma, Jennifer Brand, Hazel Quek, Pooja Venkat, Chelsea Mayoh, Aisling O'Connor, Katerina I. Leonova, Natalia Issaeva, Laura D. Gamble, Anahid Ehteda, Rachael Terry, Angelika Bongers, Emma Ronca, Mawar Karsa, Ruby Pandher, Jayne Murray, Klaartje Somers, and Lin Xiao
- Abstract
Supplementary Figures S1-S16
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Tables 1-5 from Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Michelle Haber, Murray D. Norris, Katerina V. Gurova, Andrei V. Gudkov, David S. Ziegler, Anthony J. Cesare, Ricky W. Johnstone, Glenn Marshall, Jamie I. Fletcher, Michelle J. Henderson, Daniel R. Carter, Andrew J. Gifford, Alvin Kamili, Lei Zhai, Sophie Allan, Stephanie Alfred, Georgina Eden, Adam Kearns, Erin Mosmann, Jessica A. Pettitt, Frances K. Kusuma, Jennifer Brand, Hazel Quek, Pooja Venkat, Chelsea Mayoh, Aisling O'Connor, Katerina I. Leonova, Natalia Issaeva, Laura D. Gamble, Anahid Ehteda, Rachael Terry, Angelika Bongers, Emma Ronca, Mawar Karsa, Ruby Pandher, Jayne Murray, Klaartje Somers, and Lin Xiao
- Abstract
Supplementary Tables S1-S5
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 2 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 2 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 6 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 6 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Figure 4 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 4 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 5 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 5 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 3 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 3 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure Legends 1-6 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure Legends 1-6 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
25. Data from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric muscle cancer, which arrested during the process of skeletal muscle differentiation. In muscle myoblast cells, ectopic expression of the histone H3 lysine 9 (H3K9) methytransferase KMT1A blocks differentiation by repressing a myogenic gene expression program. In this study, we tested the hypothesis that activation of a KMT1A-mediated program of transcriptional repression prevents ARMS cells from differentiating. We investigated whether KMT1A represses the expression of differentiation-associated genes in ARMS cells, thereby blocking muscle differentiation. Our results show that expression of KMT1A is induced in human ARMS cancer cell lines when cultured under differentiation-permissible conditions. shRNA-mediated knockdown of KMT1A decreased anchorage dependent and independent cell proliferation and tumor xenograft growth, increased expression of differentiation-associated genes, and promoted the appearance of a terminally differentiated-like phenotype. Finally, shRNA-directed KMT1A knockdown restored the impaired transcriptional activity of the myogenic regulator MyoD. Together, our results suggested that high levels of KMT1A in ARMS cells under differentiation conditions impairs MyoD function, thereby arresting myogenic differentiation in these tumor cells. Thus, targeting KMT1A may be a novel strategy for the treatment of this disease. Cancer Res; 71(11); 3921–31. ©2011 AACR.
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Table 1, Supplementary Figures 1-5 from Small-Molecule Multidrug Resistance–Associated Protein 1 Inhibitor Reversan Increases the Therapeutic Index of Chemotherapy in Mouse Models of Neuroblastoma
- Author
-
Michelle Haber, Andrei V. Gudkov, Murray D. Norris, Glenn M. Marshall, Alan C. Sartorelli, Katerina V. Gurova, Pavel G. Komarov, Nadezhda Isachenko, Andrei Purmal, Janice Smith, Claudia Flemming, Chengyuan Xue, Anatoly Prokvolit, Marina Pajic, Jayne Murray, Fujiko Watt, and Catherine A. Burkhart
- Abstract
Supplementary Table 1, Supplementary Figures 1-5 from Small-Molecule Multidrug Resistance–Associated Protein 1 Inhibitor Reversan Increases the Therapeutic Index of Chemotherapy in Mouse Models of Neuroblastoma
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 1 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Author
-
Asoke K. Mal, Andrei V. Gudkov, Mathivanan Jothi, and Min-Hyung Lee
- Abstract
Supplementary Figure 1 from Histone Methyltransferase KMT1A Restrains Entry of Alveolar Rhabdomyosarcoma Cells into a Myogenic Differentiated State
- Published
- 2023
- Full Text
- View/download PDF
28. Cancer relevance of circulating antibodies against LINE-1 antigens in humans
- Author
-
Alexandra V. Vylegzhanina, Ivan A. Bespalov, Ksenia A. Novototskaya-Vlasova, Brandon M. Hall, Anatoli S. Gleiberman, Han Yu, Olga V. Leontieva, Katerina I. Leonova, Oleg V. Kurnasov, Andrei L. Osterman, Grace K. Dy, Alexey A. Komissarov, Elena Vasilieva, Jeff Gehlhausen, Akiko Iwasaki, Christine B. Ambrosone, Takemasa Tsuji, Junko Matsuzaki, Kunle Odunsi, Ekaterina L. Andrianova, and Andrei V. Gudkov
- Abstract
LINE-1 (L1), the most abundant family of autonomous retrotransposons occupying over 17% of human DNA, is epigenetically silenced in normal tissues but frequently derepressed in cancer, suggesting that L1-encoded proteins may act as tumor-associated antigens recognized by the immune system. Here, we established an immunoassay for detecting circulating autoantibodies against L1 proteins in human blood. Using this assay in >3,000 individuals with or without cancer, we observed significantly higher IgG titers against L1-encoded ORF1p and ORF2p in patients with lung, pancreatic, ovarian, esophageal, and liver cancers compared to healthy individuals. Remarkably, elevated levels of anti-ORF1p-reactive IgG were observed in cancer patients with disease stages 1 and 2, indicating that immune response to L1 antigens can occur at early phases of carcinogenesis. We conclude that the antibody response against L1 antigens could contribute to the diagnosis and determination of immunoreactivity of tumors among cancer types that frequently escape early detection.
- Published
- 2023
- Full Text
- View/download PDF
29. Inflammatory response to retrotransposons drives tumor drug resistance that can be prevented by reverse transcriptase inhibitors
- Author
-
Ksenia A. Novototskaya-Vlasova, Nickolay S. Neznanov, Ivan Molodtsov, Brandon M. Hall, Mairead Commane, Anatoli S. Gleiberman, Jayne Murray, Michelle Haber, Murray D. Norris, Katerina I. Leonova, and Andrei V. Gudkov
- Subjects
Mice ,Multidisciplinary ,Long Interspersed Nucleotide Elements ,Retroelements ,Drug Resistance, Neoplasm ,NF-kappa B ,Animals ,Reverse Transcriptase Inhibitors - Abstract
Activation of endogenous retrotransposons frequently occurs in cancer cells and contributes to tumor genomic instability. To test whether inhibition of retrotranspositions has an anticancer effect, we used treatment with the nucleoside reverse transcriptase inhibitor (NRTI) stavudine (STV) in mouse cancer models, MMTV-HER2/Neu and Th-MYCN, that spontaneously develop breast cancer and neuroblastoma, respectively. In both cases, STV in drinking water did not affect tumor incidence nor demonstrate direct antitumor effects. However, STV dramatically extended progression-free survival in both models following an initial complete response to chemotherapy. To approach the mechanism underlying this phenomenon, we analyzed the effect of NRTI on the selection of treatment-resistant variants in tumor cells in culture. Cultivation of mouse breast carcinoma 4T1 in the presence of STV dramatically reduced the frequency of cells capable of surviving treatment with anticancer drugs. Global transcriptome analysis demonstrated that the acquisition of drug resistance by 4T1 cells was accompanied by an increase in the constitutive activity of interferon type I and NF-κB pathways and an elevated expression of LINE-1 elements, which are known to induce inflammatory responses via their products of reverse transcription. Treatment with NRTI reduced NF-κB activity and reverted drug resistance. Furthermore, the inducible expression of LINE-1 stimulated inflammatory response and increased the frequency of drug-resistant variants in a tumor cell population. These results indicate a mechanism by which retrotransposon desilencing can stimulate tumor cell survival during treatment and suggest reverse transcriptase inhibition as a potential therapeutic approach for targeting the development of drug-resistant cancers.
- Published
- 2022
30. Structural dissection of sequence recognition and catalytic mechanism of human LINE-1 endonuclease
- Author
-
Max Totrov, Denis N. Kazyulkin, Lioubov G. Korotchkina, Sergey Korolev, Andrei V. Gudkov, and Ian J. Miller
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,AcademicSubjects/SCI00010 ,Genetic Vectors ,Gene Expression ,Retrotransposon ,Computational biology ,Crystallography, X-Ray ,Genomic Instability ,Substrate Specificity ,Endonuclease ,chemistry.chemical_compound ,Scissile bond ,Structural Biology ,Genetics ,Consensus sequence ,DNA-(Apurinic or Apyrimidinic Site) Lyase ,Escherichia coli ,Deoxyribonuclease I ,Humans ,AP site ,Protein Interaction Domains and Motifs ,Cloning, Molecular ,DNA Cleavage ,Binding Sites ,biology ,Base Sequence ,Genome, Human ,DNA ,Recombinant Proteins ,chemistry ,biology.protein ,Nucleic Acid Conformation ,Thermodynamics ,Human genome ,Protein Conformation, beta-Strand ,Primer (molecular biology) ,Protein Binding - Abstract
Long interspersed nuclear element-1 (L1) is an autonomous non-LTR retrotransposon comprising ∼20% of the human genome. L1 self-propagation causes genomic instability and is strongly associated with aging, cancer and other diseases. The endonuclease domain of L1’s ORFp2 protein (L1-EN) initiates de novo L1 integration by nicking the consensus sequence 5′-TTTTT/AA-3′. In contrast, related nucleases including structurally conserved apurinic/apyrimidinic endonuclease 1 (APE1) are non-sequence specific. To investigate mechanisms underlying sequence recognition and catalysis by L1-EN, we solved crystal structures of L1-EN complexed with DNA substrates. This showed that conformational properties of the preferred sequence drive L1-EN’s sequence-specificity and catalysis. Unlike APE1, L1-EN does not bend the DNA helix, but rather causes ‘compression’ near the cleavage site. This provides multiple advantages for L1-EN’s role in retrotransposition including facilitating use of the nicked poly-T DNA strand as a primer for reverse transcription. We also observed two alternative conformations of the scissile bond phosphate, which allowed us to model distinct conformations for a nucleophilic attack and a transition state that are likely applicable to the entire family of nucleases. This work adds to our mechanistic understanding of L1-EN and related nucleases and should facilitate development of L1-EN inhibitors as potential anticancer and antiaging therapeutics., Graphical Abstract Graphical AbstractMechanism of DNA sequence recognition and cleavage by L1-EN endonuclease domain during initiation of the LINE-1 retrotransposon target-primed reverse transcription.
- Published
- 2021
31. Induction of monoamine oxidase A-mediated oxidative stress and impairment of NRF2-antioxidant defence response by polyphenol-rich fraction of Bergenia ligulata sensitizes prostate cancer cells in vitro and in vivo
- Author
-
Ashish Bhattacharjee, Narayan Chandra Mandal, Naibedya Dutta, Andrei V. Gudkov, Lyudmila G. Burdelya, Gopal C. Kundu, Pinaki Banerjee, Srabani Pal, V. Ravichandiran, Mahadeb Pal, Hossainoor Rahaman Sareng, Suvranil Ghosh, Prachi Kapse, and Rahul L. Gajbhiye
- Subjects
Male ,0301 basic medicine ,Programmed cell death ,NF-E2-Related Factor 2 ,Mice, SCID ,Pharmacology ,urologic and male genital diseases ,medicine.disease_cause ,Biochemistry ,Antioxidants ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Mice, Inbred NOD ,In vivo ,Physiology (medical) ,LNCaP ,Splenocyte ,medicine ,Animals ,Humans ,Bergenia ligulata ,Monoamine Oxidase ,Glycogen Synthase Kinase 3 beta ,biology ,Chemistry ,Polyphenols ,Prostatic Neoplasms ,biology.organism_classification ,Oxidative Stress ,030104 developmental biology ,biology.protein ,Monoamine oxidase A ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Prostate cancer (PCa) is a major cause of mortality and morbidity in men. Available therapies yield limited outcome. We explored anti-PCa activity in a polyphenol-rich fraction of Bergenia ligulata (PFBL), a plant used in Indian traditional and folk medicine for its anti-inflammatory and antineoplastic properties. PFBL constituted of about fifteen different compounds as per LCMS analysis induced apoptotic death in both androgen-dependent LNCaP and androgen-refractory PC3 and DU145 cells with little effect on NKE and WI38 cells. Further investigation revealed that PFBL mediates its function through upregulating ROS production by enhanced catalytic activity of Monoamine oxidase A (MAO-A). Notably, the differential inactivation of NRF2-antioxidant response pathway by PFBL resulted in death in PC3 versus NKE cells involving GSK-3β activity facilitated by AKT inhibition. PFBL efficiently reduced the PC3-tumor xenograft in NOD-SCID mice alone and in synergy with Paclitaxel. Tumor tissues in PFBL-treated mice showed upregulation of similar mechanism of cell death as observed in isolated PC3 cells i.e., elevation of MAO-A catalytic activity, ROS production accompanied by activation of β-TrCP-GSK-3β axis of NRF2 degradation. Blood counts, liver, and splenocyte sensitivity analyses justified the PFBL safety in the healthy mice. To our knowledge this is the first report of an activity that crippled NRF2 activation both in vitro and in vivo in response to MAO-A activation. Results of this study suggest the development of a novel treatment protocol utilizing PFBL to improve therapeutic outcome for patients with aggressive PCa which claims hundreds of thousands of lives each year.
- Published
- 2021
- Full Text
- View/download PDF
32. Dual Targeting of Chromatin Stability By The Curaxin CBL0137 and Histone Deacetylase Inhibitor Panobinostat Shows Significant Preclinical Efficacy in Neuroblastoma
- Author
-
Laura D. Gamble, Ricky W. Johnstone, Klaartje Somers, Rachael L. Terry, Glenn M. Marshall, Andrei V. Gudkov, Sophie Allan, Georgina L. Eden, Jamie I. Fletcher, Frances K. Kusuma, Stephanie Alfred, Mawar Karsa, Adam Kearns, Michelle J. Henderson, Michelle Haber, Angelika Bongers, Katerina Gurova, Lin Xiao, Anahid Ehteda, Lei Zhai, Erin Mosmann, Katerina I. Leonova, Natalia Issaeva, Pooja Venkat, Aisling O'Connor, Alvin Kamili, David S. Ziegler, Ruby Pandher, Daniel R. Carter, Hazel Quek, Andrew J. Gifford, Jayne Murray, Chelsea Mayoh, Jessica A. Pettitt, Murray D. Norris, Jennifer Brand, Emma Ronca, and Anthony J. Cesare
- Subjects
Cancer Research ,medicine.drug_class ,Carbazoles ,Drug Evaluation, Preclinical ,Article ,Mice ,Neuroblastoma ,chemistry.chemical_compound ,Panobinostat ,Tumor Cells, Cultured ,medicine ,Animals ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,biology ,Chemistry ,Histone deacetylase inhibitor ,Cancer ,medicine.disease ,Chromatin ,Histone Deacetylase Inhibitors ,Drug Combinations ,Histone ,Oncology ,Cancer cell ,biology.protein ,Cancer research ,Histone deacetylase - Abstract
Purpose: We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma. Experimental Design: The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction. Results: The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination. Conclusions: The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.
- Published
- 2021
- Full Text
- View/download PDF
33. A deimmunized and pharmacologically optimized Toll-like receptor 5 agonist for therapeutic applications
- Author
-
Craig M. Brackett, Catherine Burkhart, Ilia Toshkov, Andrei L. Osterman, Ivan A. Bespalov, Andrei Purmal, Vadim L. Mett, Andrei V. Gudkov, Lyudmila G. Burdelya, Yakov N. Kogan, Anatoli S. Gleiberman, Ekaterina L. Andrianova, Ivan Molodtsov, and Oleg V. Kurnasov
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,QH301-705.5 ,medicine.medical_treatment ,Medicine (miscellaneous) ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,Recombinant protein therapy ,03 medical and health sciences ,Applied immunology ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Cell Line, Tumor ,medicine ,Humans ,Biology (General) ,Toll-like receptor ,business.industry ,Immunogenicity ,Toll-Like Receptor 5 ,HEK293 Cells ,030104 developmental biology ,TLR5 ,030220 oncology & carcinogenesis ,Cancer research ,Peptides ,General Agricultural and Biological Sciences ,business ,Signal Transduction ,Entolimod - Abstract
The Toll-like receptor 5 (TLR5) agonist entolimod, a derivative of Salmonella flagellin, has therapeutic potential for several indications including radioprotection and cancer immunotherapy. However, in Phase 1 human studies, entolimod induced a rapid neutralizing immune response, presumably due to immune memory from prior exposure to flagellated enterobacteria. To enable multi-dose applications, we used structure-guided reengineering to develop a next-generation, substantially deimmunized entolimod variant, GP532. GP532 induces TLR5-dependent NF-κB activation like entolimod but is smaller and has mutations eliminating an inflammasome-activating domain and key B- and T-cell epitopes. GP532 is resistant to human entolimod-neutralizing antibodies and shows reduced de novo immunogenicity. GP532 also has improved bioavailability, a stronger effect on key cytokine biomarkers, and a longer-lasting effect on NF-κB. Like entolimod, GP532 demonstrated potent prophylactic and therapeutic efficacy in mouse models of radiation-induced death and tissue damage. These results establish GP532 as an optimized TLR5 agonist suitable for multi-dose therapies and for patients with high titers of preexisting flagellin-neutralizing antibodies., Mett et al. describe development of GP532, a substantially deimmunized derivative of Toll-like receptor 5 (TLR5) agonist entolimod. GP532 has mutations eliminating key B- and T-cell epitopes and an inflammasome-activating domain yet remains a potent NF-κB activator with biological effects similar to entolimod. Thus, GP532 is suitable for multi-dose TLR5-targeting therapies and patients with high titers of preexisting flagellin-neutralizing antibodies.
- Published
- 2021
34. Superior cancer preventive efficacy of low versus high dose of mTOR inhibitor in a mouse model of prostate cancer
- Author
-
Karen K. Kuropatwinski, Olga V. Chernova, Bryan Gillard, Anatoli S. Gleiberman, Michael T. Moser, Michelle Wrobel, Ilia Toshkov, Ellen Karasik, Andrei V. Gudkov, Ekaterina L. Andrianova, Barbara A. Foster, and Marina P. Antoch
- Subjects
PTEN ,Cancer prevention ,biology ,business.industry ,rapamycin ,Cancer Model ,Cancer ,medicine.disease ,prostate cancer ,Prostate cancer ,Oncology ,prevention ,Cancer research ,biology.protein ,mTOR ,Medicine ,business ,Energy source ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Research Paper - Abstract
// Marina P. Antoch 1 , Michelle Wrobel 2 , Bryan Gillard 1 , Karen K. Kuropatwinski 1 , Ilia Toshkov 2 , Anatoli S. Gleiberman 2 , Ellen Karasik 1 , Michael T. Moser 1 , Barbara A. Foster 1 , Ekaterina L. Andrianova 2 , Olga V. Chernova 2 and Andrei V. Gudkov 3 1 Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA 2 Everon Biosciences, Inc., Buffalo, NY, USA 3 Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA Correspondence to: Marina P. Antoch, email: marina.antoch@roswellpark.org Keywords: prostate cancer; rapamycin; prevention; mTOR; PTEN Received: February 21, 2020 Accepted: March 14, 2020 Published: April 14, 2020 ABSTRACT The mechanistic target of rapamycin (mTOR) is a PI3K-related kinase that regulates cell growth, proliferation and survival in response to the availability of energy sources and growth factors. Cancer development and progression is often associated with constitutive activation of the mTOR pathway, thus justifying mTOR inhibition as a promising approach to cancer treatment and prevention. However, development of previous rapamycin analogues has been complicated by their induction of adverse side effects and variable efficacy. Since mTOR pathway regulation involves multiple feedback mechanisms that may be differentially activated depending on the degree of mTOR inhibition, we investigated whether rapamycin dosing could be adjusted to achieve chemopreventive efficacy without side effects. Thus, we tested the efficacy of two doses of a novel, highly bioavailable nanoformulation of rapamycin, Rapatar, in a mouse prostate cancer model (male mice with prostate epithelium-specific Pten -knockout). We found that the highest efficacy was achieved by the lowest dose of Rapatar used in the study. While both doses tested were equally effective in suppressing proliferation of prostate epithelial cells, higher dose resulted in activation of feedback circuits that reduced the drug’s tumor preventive efficacy. These results demonstrate that low doses of highly bioavailable mTOR inhibitor, Rapatar, may provide safe and effective cancer prevention.
- Published
- 2020
35. Development of infrastructure for a systemic multidisciplinary approach to study aging in retired sled dogs
- Author
-
Daria Fleyshman, Ekaterina L. Andrianova, Andrei V. Gudkov, Natasha J. Olby, Joseph J. Wakshlag, Heather J. Huson, John P. Loftus, and Leonid Brodsky
- Subjects
Senescence ,Gerontology ,Aging ,senescence ,Population ,Longevity ,canine ,frailty ,healthspan ,Time frame ,Cognition ,Dogs ,Multidisciplinary approach ,Artificial Intelligence ,Medicine ,Animals ,Humans ,Cognitive decline ,education ,education.field_of_study ,Genome ,business.industry ,Cell Biology ,Disease Models, Animal ,Immune System ,Research Perspective ,business - Abstract
Canines represent a valuable model for mammalian aging studies as large animals with short lifespans, allowing longitudinal analyses within a reasonable time frame. Moreover, they develop a spectrum of aging-related diseases resembling that of humans, are exposed to similar environments, and have been reasonably well studied in terms of physiology and genetics. To overcome substantial variables that complicate studies of privately-owned household dogs, we have focused on a more uniform population composed of retired Alaskan sled dogs that shared similar lifestyles, including exposure to natural stresses, and are less prone to breed-specific biases than a pure breed population. To reduce variability even further, we have collected a population of 103 retired (8-11 years-old) sled dogs from multiple North American kennels in a specialized research facility named Vaika. Vaika dogs are maintained under standardized conditions with professional veterinary care and participate in a multidisciplinary program to assess the longitudinal dynamics of aging. The established Vaika infrastructure enables periodic gathering of quantitative data reflecting physical, physiological, immunological, neurological, and cognitive decline, as well as monitoring of aging-associated genetic and epigenetic alterations occurring in somatic cells. In addition, we assess the development of age-related diseases such as arthritis and cancer. In-depth data analysis, including artificial intelligence-based approaches, will build a comprehensive, integrated model of canine aging and potentially identify aging biomarkers that will allow use of this model for future testing of antiaging therapies.
- Published
- 2021
36. Signaling through TLR5 mitigates lethal radiation damage by neutrophil-dependent release of MMP-9
- Author
-
Lyudmila G. Burdelya, Craig M. Brackett, Kellee F. Greene, Alyssa R Aldrich, Andrei V. Gudkov, Scott I. Abrams, Nicholas H Trageser, Ivan Molodtsov, Srabani Pal, and Bojidar M. Kandar
- Subjects
Agonist ,Cancer Research ,medicine.drug_class ,Neutrophils ,Immunology ,Article ,Ionizing radiation ,Cellular and Molecular Neuroscience ,Medicine ,Regeneration ,Acute inflammation ,RC254-282 ,QH573-671 ,business.industry ,Acute Radiation Syndrome ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell Biology ,Haematopoiesis ,medicine.anatomical_structure ,Apoptosis ,TLR5 ,Cancer research ,Bone marrow ,business ,Cytology ,Entolimod - Abstract
Acute radiation syndrome (ARS) is a major cause of lethality following radiation disasters. A TLR5 agonist, entolimod, is among the most powerful experimental radiation countermeasures and shows efficacy in rodents and non-human primates as a prophylactic (radioprotection) and treatment (radiomitigation) modality. While the prophylactic activity of entolimod has been connected to the suppression of radiation-induced apoptosis, the mechanism by which entolimod functions as a radiomitigator remains poorly understood. Uncovering this mechanism has significant and broad-reaching implications for the clinical development and improvement of TLR5 agonists for use as an effective radiation countermeasure in scenarios of mass casualty resulting from accidental exposure to ionizing radiation. Here, we demonstrate that in contrast to radioprotection, neutrophils are essential for the radiomitigative activity of entolimod in a mouse model of lethal ARS. Neutrophils express functional TLR5 and rapidly exit the bone marrow (BM), accumulate in solid tissues, and release MMP-9 following TLR5 stimulation which is accompanied by an increase in the number of active hematopoietic pluripotent precursors (HPPs) in the BM. Importantly, recombinant MMP-9 by itself has radiomitigative activity and, in the absence of neutrophils, accelerates the recovery of the hematopoietic system. Unveiling this novel TLR5-neutrophil-MMP-9 axis of radiomitigation opens new opportunities for the development of efficacious radiation countermeasures to treat ARS following accidental radiation disasters.
- Published
- 2021
37. Latest advances in aging research and drug discovery
- Author
-
Daniela Bakula, Alex Zhavoronkov, Nir Barzilai, Anastasia Georgievskaya, Andrea Ablasser, Martin Immanuel Bittner, Alexander Tyshkovskiy, Martin Borch Jensen, Morten Scheibye-Knudsen, Ana Martin-Villalba, Unmesh Lal, Quentin Vanhaelen, Cornelis F. Calkhoven, Adriano Aguzzi, Jerome N. Feige, Alexey Moskalev, Andrei V. Gudkov, Danica Chen, Aubrey de Grey, Michael A. Petr, Ivan V. Ozerov, David C. Rubinsztein, Adam Antebi, Tyler Golato, Matt Kaeberlein, Thorsten Hoppe, Pekka Katajisto, Vadim N. Gladyshev, Brian K. Kennedy, Centre of Excellence in Stem Cell Metabolism, Helsinki Institute of Life Science HiLIFE, and Stem Cell Aging Leukemia and Lymphoma (SALL)
- Subjects
0301 basic medicine ,Artificial intelligence ,Aging ,Population ageing ,Drug Industry ,PROTEOSTASIS ,Psychological intervention ,translation ,Meeting Report ,Selective inhibition ,Business model ,Exhibition ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Humans ,LONGEVITY ,Healthy aging ,Pharmaceutical industry ,selective-inhibition ,Drug discovery ,business.industry ,Research ,Opinion leadership ,Cell Biology ,artificial intelligence ,3. Good health ,030104 developmental biology ,Engineering ethics ,3111 Biomedicine ,metformin ,business ,030217 neurology & neurosurgery - Abstract
An increasing aging population poses a significant challenge to societies worldwide. A better understanding of the molecular, cellular, organ, tissue, physiological, psychological, and even sociological changes that occur with aging is needed in order to treat age-associated diseases. The field of aging research is rapidly expanding with multiple advances transpiring in many previously disconnected areas. Several major pharmaceutical, biotechnology, and consumer companies made aging research a priority and are building internal expertise, integrating aging research into traditional business models and exploring new go-to-market strategies. Many of these efforts are spearheaded by the latest advances in artificial intelligence, namely deep learning, including generative and reinforcement learning. To facilitate these trends, the Center for Healthy Aging at the University of Copenhagen and Insilico Medicine are building a community of Key Opinion Leaders (KOLs) in these areas and launched the annual conference series titled "Aging Research and Drug Discovery (ARDD)" held in the capital of the pharmaceutical industry, Basel, Switzerland (www.agingpharma.org). This ARDD collection contains summaries from the 6th annual meeting that explored aging mechanisms and new interventions in age-associated diseases. The 7th annual ARDD exhibition will transpire 2nd-4th of September, 2020, in Basel.
- Published
- 2019
- Full Text
- View/download PDF
38. Potent antileukemic activity of curaxin CBL0137 against MLL‐rearranged leukemia
- Author
-
Carol Wadham, Shiloh Middlemiss, Michelle J. Henderson, Richard B. Lock, Murray D. Norris, Angelika Bongers, Katerina Gurova, Chelsea Mayoh, Mawar Karsa, Andrei V. Gudkov, Angelika Kosciolek, Michelle Haber, Ursula R. Kees, Nickolay Neznanov, Rosemary Sutton, Ali El-Ayoubi, and Klaartje Somers
- Subjects
Drug ,Cancer Research ,media_common.quotation_subject ,Carbazoles ,Antineoplastic Agents ,Apoptosis ,Chromosomal translocation ,Kaplan-Meier Estimate ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Interferon ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Medicine ,neoplasms ,media_common ,Gene Rearrangement ,business.industry ,Gene Expression Profiling ,High Mobility Group Proteins ,Myeloid leukemia ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Leukemia, Biphenotypic, Acute ,Chromatin ,DNA-Binding Proteins ,Disease Models, Animal ,Leukemia ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Transcriptional Elongation Factors ,Tumor Suppressor Protein p53 ,Transcriptome ,business ,Myeloid-Lymphoid Leukemia Protein ,Signal Transduction ,medicine.drug - Abstract
Around 10% of acute leukemias harbor a rearrangement of the MLL/KMT2A gene, and the presence of this translocation results in a highly aggressive, therapy-resistant leukemia subtype with survival rates below 50%. There is a high unmet need to identify safer and more potent therapies for MLL-rearranged (MLL-r) leukemia that can be combined with established chemotherapeutics to decrease treatment-related toxicities. The curaxin, CBL0137, has demonstrated nongenotoxic anticancer and chemopotentiating effects in a number of preclinical cancer models and is currently in adult Phase I clinical trials for solid tumors and hematological malignancies. The aim of our study was to investigate whether CBL0137 has potential as a therapeutic and chemopotentiating compound in MLL-r leukemia through a comprehensive analysis of its efficacy in preclinical models of the disease. CBL0137 decreased the viability of a panel of MLL-r leukemia cell lines (n = 12) and xenograft cells derived from patients with MLL-r acute lymphoblastic leukemia (ALL, n = 3) in vitro with submicromolar IC50s. The small molecule drug was well-tolerated in vivo and significantly reduced leukemia burden in a subcutaneous MV4;11 MLL-r acute myeloid leukemia model and in patient-derived xenograft models of MLL-r ALL (n = 5). The in vivo efficacy of standard of care drugs used in remission induction for pediatric ALL was also potentiated by CBL0137. CBL0137 exerted its anticancer effect by trapping Facilitator of Chromatin Transcription (FACT) into chromatin, activating the p53 pathway and inducing an Interferon response. Our findings support further preclinical evaluation of CBL0137 as a new approach for the treatment of MLL-r leukemia.
- Published
- 2019
- Full Text
- View/download PDF
39. Cells exhibiting strong p16 INK4a promoter activation in vivo display features of senescence
- Author
-
Brandon M. Hall, Jessica A. Sorrentino, Jie-Yu Liu, Garrett A. Sessions, Brian O. Diekman, Joel S. Parker, Janakiraman Krishnamurthy, George P. Souroullas, Andrei V. Gudkov, and Norman E. Sharpless
- Subjects
Senescence ,0303 health sciences ,Messenger RNA ,Multidisciplinary ,Cell growth ,Endogeny ,Inflammation ,Biology ,Phenotype ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Serial passage ,030220 oncology & carcinogenesis ,medicine ,medicine.symptom ,neoplasms ,030304 developmental biology - Abstract
The activation of cellular senescence throughout the lifespan promotes tumor suppression, whereas the persistence of senescent cells contributes to aspects of aging. This theory has been limited, however, by an inability to identify and isolate individual senescent cells within an intact organism. Toward that end, we generated a murine reporter strain by “knocking-in” a fluorochrome, tandem-dimer Tomato (tdTom), into exon 1α of the p16INK4a locus. We used this allele (p16tdTom) for the enumeration, isolation, and characterization of individual p16INK4a-expressing cells (tdTom+). The half-life of the knocked-in transcript was shorter than that of the endogenous p16INK4a mRNA, and therefore reporter expression better correlated with p16INK4a promoter activation than p16INK4a transcript abundance. The frequency of tdTom+ cells increased with serial passage in cultured murine embryo fibroblasts from p16tdTom/+ mice. In adult mice, tdTom+ cells could be readily detected at low frequency in many tissues, and the frequency of these cells increased with aging. Using an in vivo model of peritoneal inflammation, we compared the phenotype of cells with or without activation of p16INK4a and found that tdTom+ macrophages exhibited some features of senescence, including reduced proliferation, senescence-associated β-galactosidase (SA-β-gal) activation, and increased mRNA expression of a subset of transcripts encoding factors involved in SA-secretory phenotype (SASP). These results indicate that cells harboring activation of the p16INK4a promoter accumulate with aging and inflammation in vivo, and display characteristics of senescence.
- Published
- 2019
- Full Text
- View/download PDF
40. Resistance of bone marrow stroma to genotoxic preconditioning is determined by p53
- Author
-
Kellee F. Greene, Anatoli S. Gleiberman, Alexander N. Shakhov, Elena A. Komarova, Natalia Fedtsova, Evguenia Strom, Craig M. Brackett, Andrei V. Gudkov, Ivan Molodtsov, and Liliya R. Novototskaya
- Subjects
Cancer Research ,Stromal cell ,Immunology ,Article ,Cellular and Molecular Neuroscience ,Mice ,Stroma ,Bone Marrow ,medicine ,Animals ,Mitotic catastrophe ,Cell Proliferation ,QH573-671 ,Chemistry ,Haematopoietic stem cells ,Mesenchymal stem cell ,Cell Biology ,Total body irradiation ,Hematopoietic Stem Cells ,Stem-cell research ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Cancer research ,Bone marrow ,Tumor Suppressor Protein p53 ,Cytology - Abstract
Transplantation of bone marrow (BM) is made possible by the differential sensitivity of its stromal and hematopoietic components to preconditioning by radiation and/or chemotherapeutic drugs. These genotoxic treatments eliminate host hematopoietic precursors by inducing p53-mediated apoptosis but keep the stromal niche sufficiently intact for the engraftment of donor hematopoietic cells. We found that p53-null mice cannot be rescued by BM transplantation (BMT) from even the lowest lethal dose of total body irradiation (TBI). We compared structural changes in BM stroma of mice differing in their p53 status to understand why donor BM failed to engraft in the irradiated p53-null mice. Irradiation did not affect the general structural integrity of BM stroma and induced massive expression of alpha-smooth muscle actin in mesenchymal cells followed by increased adiposity in p53 wild-type mice. In contrast, none of these events were found in p53-null mice, whose BM stroma underwent global structural damage following TBI. Similar differences in response to radiation were observed in in vitro-grown bone-adherent mesenchymal cells (BAMC): p53-null cells underwent mitotic catastrophe while p53 wild-type cells stayed arrested but viable. Supplementation with intact BAMC of either genotype enabled donor BM engraftment and significantly extended longevity of irradiated p53-null mice. Thus, successful preconditioning depends on the p53-mediated protection of cells critical for the functionality of BM stroma. Overall, this study reveals a dual positive role of p53 in BMT: it drives apoptotic death of hematopoietic cells and protects BM stromal cells essential for its functionality.
- Published
- 2021
41. Dual targeting of the epigenome via FACT complex and histone deacetylase is a potent treatment strategy for DIPG
- Author
-
Swapna Joshi, Murray D. Norris, Sandy Simon, Orazio Vittorio, Sylvie Shen, Andrei V. Gudkov, Ornella Tolhurst, Anahid Ehteda, Caitlin Ung, Jourdin R. C. Rouaen, Ruby Pandher, Aaminah Khan, Michelle Haber, Elisha Hayden, Laura Franshaw, Rebecca Lehmann, Chelsea Mayoh, Yujie Tang, Katerina Gurova, David S. Ziegler, Maria Tsoli, Federico M. Giorgi, Anjana Gopalakrishnan, Jie Liu, Chi Nam Ignatius Pang, Anne Kankean, Peter Trebilcock, Ehteda A., Simon S., Franshaw L., Giorgi F.M., Liu J., Joshi S., Rouaen J.R.C., Pang C.N.I., Pandher R., Mayoh C., Tang Y., Khan A., Ung C., Tolhurst O., Kankean A., Hayden E., Lehmann R., Shen S., Gopalakrishnan A., Trebilcock P., Gurova K., Gudkov A.V., Norris M.D., Haber M., Vittorio O., Tsoli M., and Ziegler D.S.
- Subjects
0301 basic medicine ,xenograft model ,Cell Cycle Proteins ,Retinoblastoma Protein ,Epigenesis, Genetic ,Histones ,Epigenome ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,facilitates chromatin transcription complex ,HDAC ,Panobinostat ,Brain Stem Neoplasms ,Biology (General) ,Child ,EZH2 ,Histone deacetylase inhibitor ,High Mobility Group Proteins ,Acetylation ,Drug Synergism ,Chromatin ,pediatric cancer ,DNA-Binding Proteins ,DIPG ,Transcriptional Elongation Factors ,Neuroglia ,Signal Transduction ,medicine.drug_class ,QH301-705.5 ,Primary Cell Culture ,Carbazoles ,Antineoplastic Agents ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,03 medical and health sciences ,H3K27M ,Cell Line, Tumor ,medicine ,Animals ,Humans ,anticancer therapy ,Histone H3 acetylation ,Diffuse Intrinsic Pontine Glioma ,brainstem glioma ,Survival Analysis ,Xenograft Model Antitumor Assays ,Pediatric cancer ,030104 developmental biology ,chemistry ,E2F1 ,Cancer research ,Histone deacetylase ,Tumor Suppressor Protein p53 ,E2F1 Transcription Factor ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Summary: Diffuse intrinsic pontine glioma (DIPG) is an aggressive and incurable childhood brain tumor for which new treatments are needed. CBL0137 is an anti-cancer compound developed from quinacrine that targets facilitates chromatin transcription (FACT), a chromatin remodeling complex involved in transcription, replication, and DNA repair. We show that CBL0137 displays profound cytotoxic activity against a panel of patient-derived DIPG cultures by restoring tumor suppressor TP53 and Rb activity. Moreover, in an orthotopic model of DIPG, treatment with CBL0137 significantly extends animal survival. The FACT subunit SPT16 is found to directly interact with H3.3K27M, and treatment with CBL0137 restores both histone H3 acetylation and trimethylation. Combined treatment of CBL0137 with the histone deacetylase inhibitor panobinostat leads to inhibition of the Rb/E2F1 pathway and induction of apoptosis. The combination of CBL0137 and panobinostat significantly prolongs the survival of mice bearing DIPG orthografts, suggesting a potential treatment strategy for DIPG.
- Published
- 2021
42. Stimulation of an anti-tumor immune response with 'chromatin-damaging' therapy
- Author
-
Santosh K. Patnaik, Katerina Gurova, Andrei V. Gudkov, Junko Matsuzaki, Elizabeth A. Repasky, Craig M. Brackett, Lyudmila G. Burdelya, Adekunle Odunsi, Achamaporn Punnanitinont, Anurag K. Singh, and Minhui Chen
- Subjects
Cancer Research ,T cell ,Immunology ,Carbazoles ,chemical and pharmacologic phenomena ,Apoptosis ,Mice, SCID ,Biology ,Major histocompatibility complex ,Article ,Mice ,Immune system ,Antigen ,Interferon ,medicine ,Tumor Cells, Cultured ,Immunology and Allergy ,Animals ,Humans ,Cell Proliferation ,Mice, Inbred BALB C ,Effector ,Immunity ,Acquired immune system ,Xenograft Model Antitumor Assays ,Chromatin ,Cell biology ,medicine.anatomical_structure ,Oncology ,Colonic Neoplasms ,biology.protein ,Cytokines ,Female ,Chemokines ,CD8 ,medicine.drug - Abstract
Curaxins are small molecules that bind genomic DNA and interfere with DNA-histone interactions leading to the loss of histones and decondensation of chromatin. We named this phenomenon ‘chromatin damage’. Curaxins demonstrated anti-cancer activity in multiple pre-clinical tumor models. Here, we present data which reveals, for the first time, a role for the immune system in the anti-cancer effects of curaxins. Using the lead curaxin, CBL0137, we observed elevated expression of several group of genes in CBL0137-treated tumor cells including interferon sensitive genes, MHC molecules, some embryo-specific antigens suggesting that CBL0137 increases tumor cell immunogenicity and improves recognition of tumor cells by the immune system. In support of this, we found that the anti-tumor activity of CBL0137 was reduced in immune deficient SCID mice when compared to immune competent mice. Anti-tumor activity of CBL0137was abrogated in CD8(+) T cell depleted mice but only partially lost when NK or CD4(+) T cells were depleted. Further support for a key role for the immune system in the anti-tumor activity of CBL0137 is evidenced by an increased antigen-specific effector CD8(+) T cell and natural killer cell response, and an increased ratio of effector T cells to Tregs in the tumor and spleen. CBL0137 also elevated the number of CXCR3-expressing CTLs in the tumor and the level of interferon-γ-inducible protein 10 (IP-10) in serum, suggesting IP-10/CXCR3 controls CBL0137-elicited recruitment of effector CTLs to tumors. Our collective data underscores a previously unrecognized role for both innate and adaptive immunity in the anti-tumor activity of curaxins.
- Published
- 2020
43. Immune checkpoint protein VSIG4 as a biomarker of aging in murine adipose tissue
- Author
-
David Frescas, Evguenia Strom, Tamara Tchkonia, Valeria Kogan, Brandon M. Hall, Slavoljub Vujcic, Yi Zhu, Peter Krasnov, James L. Kirkland, Olga B. Chernova, Igor Koman, Marina P. Antoch, Andrei V. Gudkov, Olga V. Leontieva, and Anatoli S. Gleiberman
- Subjects
0301 basic medicine ,VSIG4 ,Male ,Aging ,Adipose Tissue, White ,Adipose tissue ,Inflammation ,White adipose tissue ,macrophage ,Biology ,Systemic inflammation ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,immune checkpoint ,mouse ,frailty index ,Macrophages ,Cell Biology ,Immunosenescence ,Original Articles ,Immune checkpoint ,adipose tissue ,Receptors, Complement ,Mice, Inbred C57BL ,030104 developmental biology ,inflammation ,Immunology ,Biomarker (medicine) ,Original Article ,Female ,medicine.symptom ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Adipose tissue is recognized as a major source of systemic inflammation with age, driving age‐related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA‐seq analysis of ATMs from young (8‐week‐old) and healthy aged (80‐week‐old) mice. One of the genes identified, V‐set immunoglobulin‐domain‐containing 4 (VSIG4/CRIg), encodes a Mφ‐associated complement receptor and B7 family‐related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%–52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi‐parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor‐adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4‐positive Mφ contribute to immunosenescence and/or systemic age‐related deficits., Adipocytes were removed from gonadal fat (gWAT) of old and young mice, and remaining cells were fractionated into cells capable (e.g., Mφ) and incapable of phagocyting magnetic iron particles. RNA was sequenced and analyzed to identify aging‐associated macrophage‐specific transcripts. Identified candidates were validated, and VSIG4 was confirmed a macrophageal biomarker correlating with chronological and biological age.
- Published
- 2020
44. Unsupervised learning of aging principles from longitudinal data
- Author
-
Konstantin Avchaciov, Marina P. Antoch, Ekaterina L. Andrianova, Andrei E. Tarkhov, Leonid I. Menshikov, Olga Burmistrova, Andrei V. Gudkov, and Peter O. Fedichev
- Subjects
Mice ,Aging ,Multidisciplinary ,Frailty ,Longevity ,General Physics and Astronomy ,Animals ,General Chemistry ,Neural Networks, Computer ,General Biochemistry, Genetics and Molecular Biology ,Unsupervised Machine Learning - Abstract
Age is the leading risk factor for prevalent diseases and death. However, the relation between age-related physiological changes and lifespan is poorly understood. We combined analytical and machine learning tools to describe the aging process in large sets of longitudinal measurements. Assuming that aging results from a dynamic instability of the organism state, we designed a deep artificial neural network, including auto-encoder and auto-regression (AR) components. The AR model tied the dynamics of physiological state with the stochastic evolution of a single variable, the “dynamic frailty indicator” (dFI). In a subset of blood tests from the Mouse Phenome Database, dFI increased exponentially and predicted the remaining lifespan. The observation of the limiting dFI was consistent with the late-life mortality deceleration. dFI changed along with hallmarks of aging, including frailty index, molecular markers of inflammation, senescent cell accumulation, and responded to life-shortening (high-fat diet) and life-extending (rapamycin) treatments.
- Published
- 2020
45. First-in-human study of anticancer immunotherapy drug candidate mobilan: safety, pharmacokinetics and pharmacodynamics in prostate cancer patients
- Author
-
Vasily I Kazey, Natalia V Eremina, Sergey V Mishugin, Roman V Leonenkov, Vadim L. Mett, Dmitry Pushkar, and Andrei V. Gudkov
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Metastasis ,intratumor injection ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Immune system ,Prostate ,medicine ,business.industry ,Immunotherapy ,prostate cancer ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Oncology ,TLR5 ,adenoviral vector ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Cancer research ,immunotherapy ,business ,mobilan ,Research Paper - Abstract
Toll-like receptor 5 (TLR5) controls endogenous immune responses to pathogens and is a promising target for pharmacological stimulation of anti-tumor immunity. Mobilan is an innovative gene therapy agent consisting of a non-replicating bicistronic adenovirus directing constitutive expression of human Toll-like receptor 5 (TLR5) and the secreted flagellin-based TLR5 agonist, 502s. In mice, Mobilan injection into prostate tumors resulted in autocrine TLR5 signaling, immune system activation, and suppression of tumor growth and metastasis. Here we report a first-in-human placebo-controlled clinical study of Mobilan aimed at evaluating safety, tolerability, pharmacokinetics and pharmacodynamics of a single intra-prostate injection of Mobilan in early stage prostate cancer patients. Mobilan was safe and well-tolerated at all tested doses; thus, the maximum tolerated dose was not identified. Injection of Mobilan induced signs of self-resolving inflammation not present in placebo-injected patients, including transient elevation of PSA and cytokine (G-CSF, IL-6) levels, and increased lymphoid infiltration in prostate tissue. The highest dose of Mobilan (1011 viral particles) produced the best combination of safety and pharmacodynamic effects. Therefore, Mobilan is well-tolerated and induces the expected pharmacodynamic response in humans. These results support further clinical development of Mobilan as a novel immunotherapy for prostate cancer.
- Published
- 2020
46. Identification of a blood test-based biomarker of aging through deep learning of aging trajectories in large phenotypic datasets of mice
- Author
-
Konstantin Avchaciov, Marina P. Antoch, Ekaterina L. Andrianova, Andrei E. Tarkhov, Leonid I. Menshikov, Olga Burmistrova, Andrei V. Gudkov, and Peter O. Fedichev
- Subjects
0303 health sciences ,medicine.diagnostic_test ,Artificial neural network ,Dimensionality reduction ,Phenome ,Biology ,Phenotype ,03 medical and health sciences ,0302 clinical medicine ,Exponential growth ,medicine ,Biomarker (medicine) ,Blood test ,Identification (biology) ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
We proposed and characterized a novel biomarker of aging and frailty in mice trained from the large set of the most conventional, easily measured blood parameters such as Complete Blood Counts (CBC) from the open-access Mouse Phenome Database (MPD). Instead of postulating the existence of an aging clock associated with any particular subsystem of an aging organism, we assumed that aging arises cooperatively from positive feedback loops spanning across physiological compartments and leading to an organism-level instability of the underlying regulatory network. To analyze the data, we employed a deep artificial neural network including auto-encoder (AE) and auto-regression (AR) components. The AE was used for dimensionality reduction and denoising the data. The AR was used to describe the dynamics of an individual mouse’s health state by means of stochastic evolution of a single organism state variable, the “dynamic frailty index” (dFI), that is the linear combination of the latent AE features and has the meaning of the total number of regulatory abnormalities developed up to the point of the measurement or, more formally, the order parameter associated with the instability. We used neither the chronological age nor the remaining lifespan of the animals while training the model. Nevertheless, dFI fully described aging on the organism level, that is it increased exponentially with age and predicted remaining lifespan. Notably, dFI correlated strongly with multiple hallmarks of aging such as physiological frailty index, indications of physical decline, molecular markers of inflammation and accumulation of senescent cells. The dynamic nature of dFI was demonstrated in mice subjected to aging acceleration by placement on a high-fat diet and aging deceleration by treatment with rapamycin.
- Published
- 2020
- Full Text
- View/download PDF
47. Dual Targeting of the Epigenome Via Facilitates Chromatin Transcription Complex (FACT) and Histone Deacetylase is a Potent Treatment Strategy for DIPG
- Author
-
Michelle Haber, Ornella Tolhurst, Murray D. Norris, Aaminah Khan, Sylvie Shen, Maria Tsoli, Jie Liu, Caitlin Ung, Orazio Vittorio, Yujie Tang, Andrei V. Gudkov, Jourdin R. C. Rouaen, Peter Trebilcock, Federico M. Giorgi, Chi Nam Ignatius Pang, Anjana Gopalakrishnan, Elisha Hayden, Swapna Joshi, Sandy Simon, David S. Ziegler, Chelsea Mayoh, Anahid Ehteda, Anne Kankean, Katerina Gurova, Ruby Pandher, and Laura Franshaw
- Subjects
chemistry.chemical_compound ,Histone ,chemistry ,biology ,Panobinostat ,EZH2 ,Cancer research ,biology.protein ,E2F1 ,Histone deacetylase ,Epigenome ,Epigenetics ,Chromatin - Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive and incurable childhood brain tumor for which new treatments are needed. A high throughput drug screen of 3600 pharmaceutical compounds found that anti-malarials, including quinacrine had potent activity against DIPG neurospheres. CBL0137 is a novel anti-cancer compound developed from quinacrine, which targets Facilitates Chromatin Transcription (FACT), a chromatin remodelling complex involved in transcription, replication, and DNA repair. We have found that CBL0137 displays profound cytotoxic activity against a panel of patient derived DIPG cultures, inhibiting cell proliferation and clonogenic potential, restoring tumor suppressor TP53 and Rb activity and inducing cell death through induction of apoptosis. Moreover, in an orthotopic model of DIPG, treatment with CBL0137 significantly extended animal survival. Histone mutations leading to the loss of histone trimethylation result in epigenetic dysregulation driving DIPG tumorigenesis. Treatment with CBL0137 targets this epigenetic defect, restoring both histone H3.3 acetylation and trimethylation and leading to tumor cell death. Combined epigenetic treatment with the histone deacetylase (HDAC) inhibitor panobinostat led to inhibition of the Rb/E2F1 pathway, and increased the enzymatic activity of enhancer of zeste homolog 2 (EZH2), leading to the restoration of H3K27 trimethylation. This combination therapy had synergistic activity against DIPG neurospheres with induction of apoptosis. Consistent with the in vitro results, the combination of CBL0137 and panobinostat significantly prolonged the survival of mice bearing DIPG orthografts suggesting a potential treatment strategy for DIPG.
- Published
- 2020
- Full Text
- View/download PDF
48. Abstract P4-04-02: A clinical-stage toll-like receptor 5 agonist, entolimod, boosts chemo-immunotherapy in pre-clinical TNBC by generating durable antitumor immunity
- Author
-
Craig M Brackett, Alyssa R Aldrich, Kellee F Greene, Bojidar Kandar, Jean Veith, Lyudmila G Burdelya, Scott I. Abrams, and Andrei V Gudkov
- Subjects
Cancer Research ,Oncology - Abstract
Triple-negative breast cancer (TNBC) accounts for 15-20% of all breast cancer diagnoses, is the most aggressive form of the disease, and has the poorest clinical outcomes. Effective or durable therapies are lacking because these tumors lack targets for therapeutic intervention and rapidly develop resistance to chemotherapy (CTx). The FDA has now approved chemo-immunotherapy (CTx-I) regimens (αPD-L1/nab-Paclitaxel, αPD-1/CTx) as standard-of-care for early-stage, locally advanced, and metastatic TNBC. Despite these advancements, overall response rates remain relatively low and survival outcomes are extended for only a few months especially in the case of locally advanced or metastatic disease. Thus, the need exists to develop additional or alternative strategies to potentially build upon and improve CTx-I protocols for TNBC. To this end, our laboratory has explored the importance of engaging the innate immune response to augment the efficacy of CTx-I. Previously, we demonstrated that toll-like receptor (TLR) 5 signaling can elicit antitumor immunity against a preclinical model of metastatic TNBC. In particular, systemic administration of the TLR5 agonist, entolimod, stimulated durable CD8+ T cell immunity against 4T1, the prototypical preclinical PD-L1+ metastatic TNBC model. Here, using primary 4T1 tumors to mirror the clinical setting of locally advanced disease, we show that entolimod boosts the efficacy of CTx-I (αPD-L1/Paclitaxel) by causing tumor regression. Moreover, these mice completely reject a subsequent tumor challenge, highlighting that the entolimod/αPD-L1/Paclitaxel combination generates durable antitumor immunity in such advanced preclinical settings. Importantly, systemically administered entolimod was shown to be safe in Phase I clinical trials cumulatively involving nearly 200 subjects in both healthy volunteers and advanced cancer patients. Thus, this work will aid in formulating a more effective CTx-I treatment regimen with potentially important clinical implications for patients with locally advanced or metastatic TNBC. Citation Format: Craig M Brackett, Alyssa R Aldrich, Kellee F Greene, Bojidar Kandar, Jean Veith, Lyudmila G Burdelya, Scott I. Abrams, Andrei V Gudkov. A clinical-stage toll-like receptor 5 agonist, entolimod, boosts chemo-immunotherapy in pre-clinical TNBC by generating durable antitumor immunity [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P4-04-02.
- Published
- 2022
- Full Text
- View/download PDF
49. Alkaloid-rich fraction of Ervatamia coronaria sensitizes colorectal cancer through modulating AMPK and mTOR signalling pathways
- Author
-
V. Ravichandiran, Ayan Mondal, Subhash C. Mandal, Andrei V. Gudkov, Deepak Bharadwaj Pemmaraju, Vinod K. Nelson, Anup Kumar Misra, Chirantan Majumder, Naibedya Dutta, Asif Ali, Suvranil Ghosh, Mahadeb Pal, Katerina Gurova, Aravind Kumar Rengan, and Chun-Tao Che
- Subjects
Male ,Cell cycle checkpoint ,Tabernaemontana ,Apoptosis ,P70-S6 Kinase 1 ,AMP-Activated Protein Kinases ,Mice ,Alkaloids ,Downregulation and upregulation ,Drug Discovery ,Autophagy ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Pharmacology ,Mice, Inbred BALB C ,Chemistry ,TOR Serine-Threonine Kinases ,AMPK ,Cell Cycle Checkpoints ,HCT116 Cells ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,Cancer research ,Colorectal Neoplasms ,HT29 Cells ,Signal Transduction - Abstract
Ethnopharmacological relevance Ervatamia coronaria, a popular garden plant in India and some other parts of the world is known traditionally for its anti-inflammatory and anti-cancer properties. The molecular bases of these functions remain poorly understood. Aim of the study Efficacies of the existing therapies for colorectal cancer (CRC) are limited by their life-threatening side effects and unaffordability. Therefore, identifying a safer, efficient, and affordable therapeutic is urgent. We studied the anti-CRC activity of an alkaloid-rich fraction of E. coronaria leaf extracts (AFE) and associated underlying mechanism. Materials and methods Activity guided solvant fractionation was adopted to identify the activity in AFE. Different cell lines, and tumor grown in syngeneic mice were used to understand the anti-CRC effect. Methodologies such as LCMS, MTT, RT-qPCR, immunoblot, immunohistochemistry were employed to understand the molecular basis of its activity. Results We showed that AFE, which carries about six major compounds, is highly toxic to colorectal cancer (CRC) cells. AFE induced cell cycle arrest at G1 phase and p21 and p27 genes, while those of CDK2, CDK-4, cyclin-D, and cyclin-E genes were downregulated in HCT116 cells. It predominantly induced apoptosis in HCT116p53+/+ cells while the HCT116p53-/- cells under the same treatment condition died by autophagy. Notably, AFE induced upregulation of AMPK phosphorylation, and inhibition of both of the mTOR complexes as indicated by inhibition of phosphorylation of S6K1, 4EBP1, and AKT. Furthermore, AFE inhibited mTOR-driven conversion of cells from reversible cell cycle arrest to senescence (geroconversion) as well as ERK activity. AFE activity was independent of ROS produced, and did not primarily target the cellular DNA or cytoskeleton. AFE also efficiently regressed CT26-derived solid tumor in Balb/c mice acting alone or in synergy with 5FU through inducing autophagy as a major mechanism of action as indicated by upregulation of Beclin 1 and phospho-AMPK, and inhibition of phospho-S6K1 levels in the tumor tissue lysates. Conclusion AFE induced CRC death through activation of both apoptotic and autophagy pathways without affecting the normal cells. This study provided a logical basis for consideration of AFE in future therapy regimen to overcome the limitations associated with existing anti-CRC chemotherapy.
- Published
- 2022
- Full Text
- View/download PDF
50. Quantitative characterization of biological age and frailty based on locomotor activity records
- Author
-
Konstantin Avchaciov, Mikhail A. Pyatnitskiy, Kristina Khodova, L. I. Menshikov, Evgeny Getmantsev, Peter O. Fedichev, Boris Zhurov, Timothy V. Pyrkov, and Andrei V. Gudkov
- Subjects
Gerontology ,Male ,0301 basic medicine ,Aging ,Multivariate analysis ,Time Factors ,Psychological intervention ,physical activity ,Locomotor activity ,0302 clinical medicine ,Risk Factors ,Medicine ,Child ,Aged, 80 and over ,Frailty ,Incidence (epidemiology) ,Age Factors ,Middle Aged ,Nutrition Surveys ,Biobank ,Natural measure ,Child, Preschool ,Principal component analysis ,Female ,Gompertz–Makeham law of mortality ,Locomotion ,Research Paper ,Adult ,UK Biobank ,Adolescent ,National Health and Nutrition Examination Survey ,Frail Elderly ,Biological age ,Gompertz function ,Physical activity ,biological clock ,Biology ,Models, Biological ,Risk Assessment ,Young Adult ,03 medical and health sciences ,Predictive Value of Tests ,Humans ,NHANES ,Set (psychology) ,Geriatric Assessment ,Simulation ,Aged ,Models, Statistical ,business.industry ,Proportional hazards model ,Type 2 Diabetes Mellitus ,Cell Biology ,health span ,Actigraphy ,United Kingdom ,United States ,030104 developmental biology ,business ,030217 neurology & neurosurgery ,Demography - Abstract
Since chronological age is not a complete and accurate indicator of organism aging, the concept of 9biological age9 has emerged as a well-accepted way to quantify the aging process in humans and laboratory animals. In this study, we performed a systematic statistical evaluation of the relationships between locomotor activity and biological age, mortality risk, and frailty using human physical activity records from the 2003-2006 National Health and Nutrition Examination Survey (NHANES) and UK BioBank (UKBB) databases. These records are from subjects ranging from 5 to 85 years old and include 7-day long continuous tracks of activity provided by wearable monitors as well as data for a comprehensive set of clinical parameters, lifestyle information and death records, thus enabling quantitative assessment of frailty and mortality. We proposed a statistical description of the locomotor activity tracks and transformed the provided time series into vectors representing individual physiological states for each participant. Using this data, we performed an unsupervised multivariate analysis and observed development and aging as a continuous trajectory consisting of distinct phases, each corresponding to subsequent human life stages. Therefore, we suggest the distance measured along this trajectory as a definition of the biological age. Consistent with the Gompertz law, mortality, estimated with the help of a proportional hazard model, was found to be an exponential function of biological age as quantified herein. However, we observed that the significant contribution of clinical frailty to mortality risk can be independent of biological age. We used the biological age and mortality models to show that some lifestyle variables, such as smoking, produce a reversible increase in all-cause mortality without a significant effect on biological age. In contrast, medical conditions, such as type 2 diabetes mellitus (T2DM) or hypertension, are associated with significant aging acceleration and a corresponding increase in mortality as well. The results of this work demonstrate that significant information relevant to aging can be extracted from human locomotor activity data and highlight the opportunity provided by explosive deployment of wearable sensors to use such information to encourage lifestyle modifications and clinical development of therapeutic interventions against the aging process.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.