300 results on '"Skorski T"'
Search Results
52. Growth factor-dependent inhibition of normal hematopoiesis by N-ras antisense oligodeoxynucleotides.
- Author
-
Skorski, T, primary, Szczylik, C, additional, Ratajczak, M Z, additional, Malaguarnera, L, additional, Gewirtz, A M, additional, and Calabretta, B, additional
- Published
- 1992
- Full Text
- View/download PDF
53. 31. Inhibition of proliferation by c-myb antisense RNA and oligodeoxynucleotidas in transformed neuroectodermal cell lines
- Author
-
Raschella, G, primary, Negreni, A, additional, Skorski, T, additional, Pucci, S, additional, Nieborowska-Skorska, M, additional, Romeo, A, additional, and Calabretta, B, additional
- Published
- 1992
- Full Text
- View/download PDF
54. 44. Growth factor-dependent inhibition of normal hematopoiesis by N-ras antisens oligodeoxynucieotides
- Author
-
Skorski, T, primary, Szczylik, C, additional, Ratajczak, MZ, additional, Malaguarnera, L, additional, Gewirtz, AM, additional, and Calabretta, B, additional
- Published
- 1992
- Full Text
- View/download PDF
55. Novel cationic amphiphiles as delivery agents for antisense oligonucleotides.
- Author
-
DeLong, R. K., Yoo, Hoon, Alahari, S. K., Fisher, M., Short, S. M., Kang, S. H., Kole, R., Janout, V., Regan, S. L., Juliano, R. L., Verma, I.M., Crooke, S.T., Roush, W., Ho, P.T., Stull, R.A., Alahari, S.K., Bennett, C.F., Skorski, T., Dean, N.M., and Zabner, J.
- Published
- 1999
- Full Text
- View/download PDF
56. Selective inhibition of leukemia cell proliferation by BCR-ABL antisense oligodeoxynucleotides.
- Author
-
Szczylik, C. and Skorski, T.
- Subjects
- *
LEUKEMIA - Abstract
Describes a study designed to determine the role of the BCR-ABL gene in the proliferation of blast cells of patients with chronic myelogenous leukemia.
- Published
- 1991
- Full Text
- View/download PDF
57. Cytochemical Characterization of Mouse L1210 Leukemia.
- Author
-
Kawiak, J., Skorski, T., Ciechanowicz, A., Zylińńska, M., Seidel, H. J., Kawalec, M., Gnatowski, B., and Czarnomska, A.
- Published
- 1988
- Full Text
- View/download PDF
58. Overexpression of the zinc finger protein MZF1 inhibits hematopoietic development from embryonic stem cells: correlation with negative regulation of CD34 and c-myb promoter activity
- Author
-
Perrotti, D, Melotti, P, Skorski, T, Casella, I, Peschle, C, and Calabretta, B
- Abstract
Zinc finger genes encode proteins that act as transcription factors. The myeloid zinc finger 1 (MZF1) gene encodes a zinc finger protein with two DNA-binding domains that recognize two distinct consensus sequences, is preferentially expressed in hematopoietic cells, and may be involved in the transcriptional regulation of hematopoiesis-specific genes. Reverse transcription-PCR analysis of human peripheral blood CD34+ cells cultured under lineage-restricted conditions demonstrated MZF1 expression during both myeloid and erythroid differentiation. Sequence analysis of the 5'-flanking region of the CD34 and c-myb genes, which are a marker of and a transcriptional factor required for hematopoietic proliferation and differentiation, respectively, revealed closely spaced MZF1 consensus binding sites found by electrophoretic mobility shift assays to interact with recombinant MZF1 protein. Transient or constitutive MZF1 expression in different cell types resulted in specific inhibition of chloramphenicol acetyltransferase activity driven by the CD34 or c-myb 5'-flanking region. To determine whether transcriptional modulation by MZF1 activity plays a role in hematopoietic differentiation, constructs containing the MZF1 cDNA under the control of different promoters were transfected into murine embryonic stem cells which, under defined in vitro culture conditions, generate colonies of multiple hematopoietic lineages. Constitutive MZF1 expression interfered with the ability of embryonic stem cells to undergo hematopoietic commitment and erythromyeloid colony formation and prevented the induced expression of CD34 and c-myb mRNAs during differentiation of these cells. These data indicate that MZF1 plays a critical role in hematopoiesis by modulating the expression of genes involved in this process.
- Published
- 1995
- Full Text
- View/download PDF
59. Oncogene-targeted antisense oligodeoxynucleotides combined with chemotherapy or immunotherapy: A new approach for tumor treatment?
- Author
-
Nieborowskaskorska, M., Nakashima, M., Mariusz Z. Ratajczak, Steplewski, Z., Calabretta, B., and Skorski, T.
- Subjects
Fusion Proteins, bcr-abl ,Down-Regulation ,Bone Marrow Cells ,DNA, Neoplasm ,Oncogenes ,Oligonucleotides, Antisense ,Polymerase Chain Reaction ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-myc ,Immunotherapy ,Leukemia BCR-ABL Positive/drug therapy ,Melanoma/drug therapy ,Oligonucleotides Antisense/therapeutic use ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Tumor Cells, Cultured ,Humans ,RNA, Messenger ,Melanoma ,Cell Division ,T-Lymphocytes, Cytotoxic - Abstract
Synthetic oligodeoxynucleotides (antisenses) complementary to bcr/abl breakpoint junction transcript on Philadelphia chromosome, or c-myb protooncogene inhibit partially the proliferation of Philadelphia positive leukemic cells (antisenses against bcr/abl and c-myb) and other tumor cells (antisenses against c-myb). This phenomenon is accompanied by specific downregulation of mRNA level of the particular gene. To develop a more effective procedure of tumor treatment the combination of low dose of cytostatic and bcr/abl or c-myb antisenses against Philadelphia chromosome positive cell line BV173, and the combination of anti-tumor cytotoxic T lymphocytes (CTL) and c-myb antisenses against melanoma cell line MM-28, were tested in vitro. Our results indicate that the combinations of conventional chemotherapeutic agent and antisense against bcr/abl or c-myb or tumor specific CTL and antisense against c-myb, are highly effective in killing of tumor cells and sparing normal cells. This creates the possibility to develop a more selective and effective treatment of neoplasia.
60. BCR/ABL ONCOGENIC KINASE INHIBITS MISMATCH REPAIR TO PROTECT FROM APOPTOSIS AND INDUCE POINT MUTATIONS
- Author
-
Stoklosa, T. S., Poplawski, T., Koptyra, M., Nieborowska-Skorska, M., Basak, G., Slupianek, A., Rayevskaya, M., Seferynska, I., Herrera, L., Blasiak, J., and Skorski, T.
61. PRE-EXISTING SOMATIC MUTATIONS IN GENES COMMONLY MUTATED IN MYELOID MALIGNANCIES (RUNX1, DNMT3A, ASXL1) MAY CONFER BAD PROGNOSIS IN CHRONIC MYELOID LEUKEMIA
- Author
-
Machnicki, M. M., Iwona Solarska, Zawada, M., Stawinski, P., Ploski, R., Seferynska, I., Sacha, T., Skorski, T., and Stoklosa, T.
62. Role of p53 in hematopoietic recovery after cytotoxic treatment
- Author
-
Wlodarski, P., Wasik, M., Ratajczak, M. Z., Sevignani, C., Hoser, G., Jerzy Kawiak, Gewirtz, A. M., Calabretta, B., and Skorski, T.
- Subjects
Mice, Knockout ,Antimetabolites ,Apoptosis ,Hematopoiesis/genetics ,Hematopoiesis ,Antimetabolites/toxicity ,Mice ,Gene Expression Regulation ,Apoptosis/genetics ,Animals ,Genes, Tumor Suppressor ,Fluorouracil ,Tumor Suppressor Protein p53 - Abstract
Prompt reconstitution of hematopoiesis after cytoreductive therapy is essential for patient recovery and may have a positive impact on long-term prognosis. We examined the role of the p53 tumor suppressor gene in hematopoietic recovery in vivo after treatment with the cytotoxic drug 5-fluorouracil (5-FU). We used p53 knock-out (p53-/-) and wild-type (p53+/+) mice injected with 5-FU as the experimental model. Analysis of the repopulation ability and clonogenic activity of hematopoietic stem cells (HSCs) and their lineage-committed descendants showed a greater number of HSCs responsible for reconstitution of lethally irradiated recipients in p53-/- bone marrow cells (BMCs) recovering after 5-FU treatment than in the corresponding p53+/+ BMCs. In post-5-FU recovering BMCs, the percentage of HSC-enriched Lin- Sca-1(+) c-Kit+ cells was about threefold higher in p53-/- than in p53+/+ cells. Although the percentage of the most primitive HSCs (Lin- Sca-1(+) c-Kit+ CD34(low/-)) did not depend on p53, the percentage of multipotential HSCs and committed progenitors (Lin- Sca-1(+) c-Kit+ CD34(high/+)) was almost fourfold higher in post-5-FU recovering p53-/- BMCs than in their p53+/+ counterparts. The pool of HSCs from 5-FU-treated p53-/- BMCs was exhausted more slowly than that from the p53+/+ population as shown in vivo using pre-spleen colony-forming unit (CFU-S) assay and in vitro using long-term culture-initiating cells (LTC-ICs) and methylcellulose replating assays. Clonogenic activity of various lineage-specific descendants was significantly higher in post-5-FU regenerating p53-/- BMCs than in p53+/+ BMCs, probably because of their increased sensitivity to growth factors. Despite all these changes and the dramatic difference in sensitivity of p53-/- and p53+/+ BMCs to 5-FU-induced apoptosis, lineage commitment and differentiation of hematopoietic progenitors appeared to be independent of p53 status. These studies suggest that suppression of p53 function facilitates hematopoietic reconstitution after cytoreductive therapy by: (1) delaying the exhaustion of the most primitive HSC pool, (2) stimulating the production of multipotential HSCs, (3) increasing the sensitivity of hematopoietic cells to growth factors, and (4) decreasing the sensitivity to apoptosis.
63. Suppression of Philadelphia leukemia cell growth in mice by bcr-abl antisense oligodeoxynucleotides
- Author
-
Skorski, T., Nieborowska-Skorska, M., Nicolaides, N. C., Szczylik, C., Patrick Iversen, Iozzo, R. V., Zon, G., and Calabretta, B.
64. Inhibition of proliferation by c-myb antisense RNA and oligodeoxynucleotides in transformed neuroectodermal cell lines
- Author
-
Raschellà, G., Negroni, A., Skorski, T., Sabina Pucci, Nieborowska-Skorska, M., Romeo, A., and Calabretta, B.
- Subjects
Transcription, Genetic ,Cell Division/drug effects ,Genetic Vectors ,Molecular Sequence Data ,Oligonucleotides ,Transfection ,Polymerase Chain Reaction ,Cell Line ,Neuroblastoma ,Genetic ,Proto-Oncogenes ,Animals ,Humans ,RNA, Antisense ,Oligonucleotides Antisense/pharmacology ,RNA, Neoplasm ,Cloning, Molecular ,Antisense ,Cell Line, Transformed ,Base Sequence ,Molecular ,Oncogenes ,Oligonucleotides, Antisense ,Oligodeoxyribonucleotides ,Transformed ,Settore MED/03 - Genetica Medica ,RNA ,Neoplasm ,Cell Division ,Transcription ,Cloning - Abstract
Transfection of a neuroblastoma cell line with expression vectors containing two different segments of human c-myb complementary DNA in antisense orientation yielded far fewer transfectant clones than did the transfection with the identical segments in sense orientation. In cell clones expressing c-myb antisense RNA, levels of the c-myb protein were down-regulated and the proliferation rate was slower than that of cells transfected with sense constructs or the untransfected parental cell line. Treatment of neuroblastoma and neuroepithelioma cell lines with a c-myb antisense oligodeoxynucleotide strongly inhibited cell growth. These data indicate a definite involvement of c-myb in the proliferation of neuroectodermal tumor cells extending the role of this protooncogene beyond the hematopoietic system. The availability of cell clones that transcribe c-myb antisense RNA provides a useful tool to study the involvement of other genes in the proliferation and differentiation of neuroblastoma cells.
65. Return of immunohematopoietic impairment of long time after murine syngeneic bone marrow transplantation
- Author
-
Skorski, T., Kawalec, M., Ratajczak, M., Szczylik, C., and Jerzy Kawiak
66. The role of c-myc protooncogene in chronic myelogenous leukemia
- Author
-
Nieborowskaskorska, M., Mariusz Z. Ratajczak, Calabretta, B., and Skorski, T.
- Subjects
Base Sequence ,Blotting, Western ,Molecular Sequence Data ,Genes, myc ,Down-Regulation ,Gene myc/genetics ,DNA, Antisense ,Leukemia Myelogenous Chronic BCR-ABL Positive/genetics ,Gene Expression Regulation, Neoplastic ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Tumor Cells, Cultured ,Humans ,Electrophoresis, Polyacrylamide Gel ,Cell Division - Abstract
c-Myc transcriptional factor encoded by c-myc protooncogene plays an important role in the regulation of cell cycle. It was also established that c-Myc is important for the transformation of fibroblasts and murine bone marrow cells induced by BCR/ABL tyrosine kinase encoded by bcr/abl oncogene localized on Philadelphia-chromosome (Ph1). The role of c-Myc in the proliferation of the leukemic cells was not known. Therefore, we examined the effect of c-Myc protein downregulation, using antisense oligodeoxynucleotides, on the growth of the BCR/ABL- dependent cell line and chronic myelogenous leukemia (CML) patients cells. Downregulation of c-Myc expression caused complete inhibition of the proliferation of BCR/ABL-dependent BV173 cell line and 50-70% inhibition of the colony formation of CML cells. These results suggests that c-Myc cooperates with BCR/ABL and is necessary for the growth of Ph1-positive leukemias.
67. Chronic myeloid leukemia--some topical issues
- Author
-
Mughal, T., Cortes, J., Cross, N. C. P., Donato, N., Hantschel, O., Jabbour, E., Kantarjian, H., Melo, J. V., Skorski, T., Silver, R. T., and Goldman, J. M.
68. GENOMIC CHARACTERIZATION OF CML AT DIAGNOSIS REVEALS PREEXISTING SOMATIC MUTATIONS THAT MAY PREDICT PROGRESSION TO BLASTIC PHASE INDEPENDENTLY OF BCR-ABL1 MUTATIONS
- Author
-
Katarzyna Borg, Barbara Pienkowska-Grela, Machnicki, M. M., Iwona Solarska, Zawada, M., Tomasz Stoklosa, Tomasz Sacha, Joanna Góra-Tybor, Ilona Seferyńska, Pruszczyk, K., Piotr Stawiński, Rafał Płoski, Skorski, T., Niesiobędzka-Krężel, J., Sawicki, W., and Joanna Kosinska
69. Pre-stimulation of the human bone marrow CD34+ cells before storage at 4 degrees C with the early acting cytokines enhance their survival and increase proliferative potential. Transplantological implications
- Author
-
Ratajczak, M. Z., Kuczynski, W. I., Skorski, T., and Ratajczak, J.
70. Oligonucleotide N3'-> P5' phosphoramidates as antisense agents
- Author
-
Gryaznov, S., Skorski, T., Cucco, C., Nieborowskaskorska, M., Chiu, C.Y., Lloyd, D., Chen, J.K., Koziolkiewicz, M., and Calabretta, B.
- Subjects
Antisense nucleic acids -- Research ,Health ,Research - Abstract
According to the authors' abstract of an article published in Nucleic Acids Research, 'Uniformly modified oligonucleotide N3'-->P5' phosphoramidates, where every 3'-oxygen is replaced by a 3'-amino group, were synthesized. These [...]
- Published
- 1996
71. Treatment of Philadelphia leukemia in severe combined immunodeficient mice by combination of cyclophosphamide and bcr/abl antisense oligodeoxynucleotides
- Author
-
Skorski, T., Nieborowskaskorska, M., Wlodarski, P., Perrotti, D., Hoser, G., Kawiak, J., Majewski, M., Christensen, L., Iozzo, R.V., and Calabretta, B.
- Subjects
Cyclophosphamide -- Health aspects ,Antisense nucleic acids -- Health aspects ,Chemotherapy -- Health aspects ,Myeloid leukemia, Philadelphia-positive -- Health aspects ,Cancer -- Chemotherapy ,Myeloid leukemia, Philadelphia-negative -- Health aspects ,Health ,Health aspects - Abstract
Skorski, T.; Nieborowskaskorska, M.; Wlodarski, P.; Perrotti, D.; Hoser, G.; Kawiak, J.; Majewski, M.; Christensen, L.; Iozzo, R.V.; Calabretta, B. 'Treatment of Philadelphia Leukemia in Severe Combined Immunodeficient Mice by [...]
- Published
- 1997
72. Leukemia treatment in severe combined immunodeficiency mice by antisense oligodeoxynucleotides targeting cooperating oncogenes
- Author
-
Skorski, T., Nieborowskaskorska, M., Campbell, K., Iozzo, R.V., Zon, G., Darzynkiewicz, Z., and Calabretta, B.
- Subjects
Antisense nucleic acids -- Health aspects ,Myeloid leukemia, Philadelphia-positive -- Care and treatment ,Oncogenes -- Physiological aspects ,Business ,Health care industry - Abstract
According to the authors' abstract of an article published in Journal of Experimental Medicine, 'Transformation of hematopoietic cells by the p210(bcr/abl) tyrosine kinase appears to require the expression of a [...]
- Published
- 1996
73. Chronic myeloid leukemia 2011: Successes, challenges, and strategies-Proceedings of the 5th annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms workshop
- Author
-
Jerald P. Radich, Tariq I. Mughal, Giovanni Martinelli, Ayalew Tefferi, Daniel J. DeAngelo, Alfonso Quintás-Cardama, Carlo Gambacorti-Passerini, Richard A. Van Etten, Farhad Ravandi, Tomasz Skorski, Mughal T.I., Radich J.P., Van Etten R.A., Quintás-Cardama A., Skorski T., Ravandi F., Deangelo D.J., Gambacorti-Passerini C., Martinelli G., Tefferi A., Mughal, T, Radich, J, Van Etten, R, Quintás Cardama, A, Skorski, T, Ravandi, F, Deangelo, D, GAMBACORTI PASSERINI, C, Martinelli, G, and Tefferi, A
- Subjects
medicine.medical_specialty ,Gene mutation ,Article ,Genomic Instability ,Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative ,Antineoplastic Agent ,chemistry.chemical_compound ,Bcr abl1 ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animal ,business.industry ,Ponatinib ,Myeloid leukemia ,Imatinib ,Hematology ,Dasatinib ,Imatinib mesylate ,chemistry ,Drug Resistance, Neoplasm ,Family medicine ,Immunology ,Drug Monitoring ,business ,Bosutinib ,CHRONIC MYELOID LEUKEMIA (CML) ,Human ,medicine.drug - Abstract
NIH Public Access Author Manuscript Am J Hematol. Author manuscript; available in PMC 2012 November 01. Published in final edited form as: Am J Hematol. 2011 September ; 86(9): 811–819. doi:10.1002/ajh.22097. $watermark-text Chronic Myeloid Leukemia 2011: Successes, challenges, and strategies – Proceedings of the 5 th Annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms workshop Tariq I Mughal 1 , Jerald P Radich 2 , Richard A. Van Etten 3 , Alfonso Quintas-Cardama 4 , Tomasz Skorski 5 , Farhad Ravandi 6 , Daniel J. DeAngelo 7 , Carlo Gambacorti-Passerini 8 , Giovanni Martinelli 9 , and Ayalew Tefferi 10 1 University of Colorado School of Medicine, Denver, USA 2 Fred Hutchinson Cancer Center, Seattle, USA 3 Tufts Medical Center, Boston, USA 4 MD Anderson Cancer Center, Houston, USA $watermark-text 5 Temple 6 MD University, Philadelphia, USA Anderson Cancer Center, Houston, USA 7 Dana Farber Cancer Center at Harvard Medical School, Boston, USA 8 University 9 Insitute 10 Mayo of Milan, Monza, Italy Le A. Seragnoli, Bologna, Italy Clinic, Rochester, MN, USA. Abstract $watermark-text This report is based on the presentations and discussions at the 5 th annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms (MPN) workshop, which took place immediately following the 52 nd American Society of Hematology (ASH) meeting in Orlando, Florida on December 7 th -8 th , 2011. Relevant data which was presented at the ASH meeting as well as all other recent publications were presented and discussed at the workshop. This report covers front-line therapies of BCR-ABL1-positive leukemias, in addition to addressing some topical biological, pre-clinical and clinical issues, such as new insights into genomic instability and resistance to tyrosine kinase inhibitors (TKIs), risk stratification and optimizing molecular monitoring. A report pertaining to the new therapies and other pertinent preclinical and clinical issues in the BCR-ABL1 negative MPNs is published separately. Introduction For patients with BCR-ABL1-positive leukemias, which comprise of all the Philadelphia (Ph) chromosome-positive and some Ph-negative leukemias, the introduction of the original tyrosine kinase inhibitor (TKI), imatinib mesylate (IM) into the clinics in 1998, resulted in being a classic therapeutic landmark (1,2). After 12 months of therapy with IM, 69% of Correspondence to: Tariq Mughal MD FRCP FACP 23655 Currant Drive, Golden, Colorado 80401, USA Tel +1 303 526 8586 tmughal911@hotmail.com.
- Published
- 2011
74. Advances in the biology and therapy of chronic myeloid leukemia: proceedings from the 6th Post-ASH International Chronic Myeloid Leukemia and Myeloproliferative Neoplasms Workshop
- Author
-
Tomasz Skorski, Michael J. Mauro, David T. Scadden, Jerald P. Radich, Ayalew Tefferi, Carlo Gambacorti-Passerini, Tariq I. Mughal, Richard A. Van Etten, Rüdiger Hehlmann, John M. Goldman, Giovanni Martinelli, Simona Soverini, Giuseppe Saglio, Catriona Jamieson, Danilo Perrotti, Van Etten, R, Mauro, M, Radich, J, Goldman, J, Saglio, G, Jamieson, C, Soverini, S, GAMBACORTI PASSERINI, C, Hehlmann, R, Martinelli, G, Perrotti, D, Scadden, D, Skorski, T, Tefferi, A, and Mughal, T
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Dasatinib ,Tyrosine kinase inhibitor ,Protein Kinase Inhibitor ,Antineoplastic Agents ,Tyrosine-kinase inhibitor ,Article ,Antineoplastic Agent ,Myelogenous ,Internal medicine ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Stem Cell Niche ,Protein Kinase Inhibitors ,Animal ,business.industry ,Myeloid leukemia ,Hematology ,Genomics ,Nilotinib ,medicine.disease ,Imatinib mesylate ,Leukemia ,Treatment Outcome ,Immunology ,Genomic ,Neoplastic Stem Cells ,Neoplastic Stem Cell ,business ,Chronic myelogenous leukemia ,Human ,medicine.drug - Abstract
Following the 53rd annual meeting of the American Society of Hematology (ASH) in San Diego in December 2011, a group of clinical and laboratory investigators convened for the 6th Post-ASH International Workshop on Chronic Myeloid Leukemia (CML) and Myeloproliferative Neoplasms (MPN). The Workshop took place on 13-14 December at the Estancia, La Jolla, California, USA. This report summarizes the most recent advances in the biology and therapy of CML that were presented at the ASH meeting and discussed at the Workshop. Preclinical studies focused on the CML stem cell and its niche, and on early results of deep sequencing of CML genomes. Clinical advances include updates on second- and third-generation tyrosine kinase inhibitors (TKIs), molecular monitoring, TKI discontinuation studies and new therapeutic agents. A report summarizing the pertinent advances in MPN has been published separately. © 2013 Informa UK, Ltd.
- Published
- 2012
75. Star wars against leukemia: attacking the clones.
- Author
-
Toma MM and Skorski T
- Subjects
- Humans, Tumor Microenvironment, Epigenesis, Genetic, Leukemia genetics, Leukemia pathology
- Abstract
Leukemia, although most likely starts as a monoclonal genetic/epigenetic anomaly, is a polyclonal disease at manifestation. This polyclonal nature results from ongoing evolutionary changes in the genome/epigenome of leukemia cells to promote their survival and proliferation advantages. We discuss here how genetic and/or epigenetic aberrations alter intracellular microenvironment in individual leukemia clones and how extracellular microenvironment selects the best fitted clones. This dynamic polyclonal composition of leukemia makes designing an effective therapy a challenging task especially because individual leukemia clones often display substantial differences in response to treatment. Here, we discuss novel therapeutic approach employing single cell multiomics to identify and eradicate all individual clones in a patient., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
76. DNA polymerase theta-mediated DNA repair is a functional dependency and therapeutic vulnerability in DNMT3A deficient leukemia cells.
- Author
-
Le BV, Vekariya U, Toma MM, Nieborowska-Skorska M, Caron MC, Gozdecka M, Haydar Z, Walsh M, Ghosh J, Vaughan-Williams E, Podszywalow-Bartnicka P, Kukuyan AM, Ziolkowska S, Hadzijusufovic E, Chandramouly G, Piwocka K, Pomerantz R, Vassiliou GS, Huntly BJ, Valent P, Bellacosa A, Masson JY, Gupta GP, Challen GA, and Skorski T
- Abstract
Myeloid malignancies carrying somatic DNMT3A mutations (DNMT3Amut) are usually resistant to standard therapy. DNMT3Amut leukemia cells accumulate toxic DNA double strand breaks (DSBs) and collapsed replication forks, rendering them dependent on DNA damage response (DDR). DNA polymerase theta (Polθ), a key element in Polθ-mediated DNA end-joining (TMEJ), is essential for survival and proliferation of DNMT3Amut leukemia cells. Polθ is overexpressed in DNMT3Amut leukemia cells due to abrogation of PARP1 PARylation-dependent UBE2O E3 ligase-mediated ubiquitination and proteasomal degradation of Polθ. In addition, PARP1-mediated recruitment of the SMARCAD1-MSH2/MSH3 repressive complex to DSBs was diminished in DNMT3Amut leukemia cells which facilitated loading of Polθ on DNA damage and promoting TMEJ and replication fork restart. Polθ inhibitors enhanced the anti-leukemic effects of mainstream drugs such as FLT3 kinase inhibitor quizartinib, cytarabine and etoposide in vitro and in mice with FLT3(ITD);DNMT3Amut leukemia. Altogether, Polθ is an attractive target in DNMT3Amut hematological malignancies.
- Published
- 2024
- Full Text
- View/download PDF
77. Polθ Inhibitor (ART558) Demonstrates a Synthetic Lethal Effect with PARP and RAD52 Inhibitors in Glioblastoma Cells.
- Author
-
Barszczewska-Pietraszek G, Czarny P, Drzewiecka M, Błaszczyk M, Radek M, Synowiec E, Wigner-Jeziorska P, Sitarek P, Szemraj J, Skorski T, and Śliwiński T
- Subjects
- Humans, Cell Line, Tumor, Cell Proliferation drug effects, DNA Polymerase theta, Apoptosis drug effects, DNA Damage drug effects, DNA-Directed DNA Polymerase metabolism, DNA-Directed DNA Polymerase genetics, Synthetic Lethal Mutations drug effects, Astrocytes drug effects, Astrocytes metabolism, Glioblastoma drug therapy, Glioblastoma pathology, Glioblastoma metabolism, Glioblastoma genetics, Rad52 DNA Repair and Recombination Protein metabolism, Rad52 DNA Repair and Recombination Protein genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Temozolomide pharmacology, Poly (ADP-Ribose) Polymerase-1 antagonists & inhibitors, Poly (ADP-Ribose) Polymerase-1 metabolism, Cell Survival drug effects
- Abstract
DNA repair proteins became the popular targets in research on cancer treatment. In our studies we hypothesized that inhibition of DNA polymerase theta (Polθ) and its combination with Poly (ADP-ribose) polymerase 1 (PARP1) or RAD52 inhibition and the alkylating drug temozolomide (TMZ) has an anticancer effect on glioblastoma cells (GBM21), whereas it has a low impact on normal human astrocytes (NHA). The effect of the compounds was assessed by analysis of cell viability, apoptosis, proliferation, DNA damage and cell cycle distribution, as well as gene expression. The main results show that Polθ inhibition causes a significant decrease in glioblastoma cell viability. It induces apoptosis, which is accompanied by a reduction in cell proliferation and DNA damage. Moreover, the effect was stronger when dual inhibition of Polθ with PARP1 or RAD52 was applied, and it is further enhanced by addition of TMZ. The impact on normal cells is much lower, especially when considering cell viability and DNA damage. In conclusion, we would like to highlight that Polθ inhibition used in combination with PARP1 or RAD52 inhibition has great potential to kill glioblastoma cells, and shows a synthetic lethal effect, while sparing normal astrocytes.
- Published
- 2024
- Full Text
- View/download PDF
78. Structural basis for a Polθ helicase small-molecule inhibitor revealed by cryo-EM.
- Author
-
Ito F, Li Z, Minakhin L, Chandramouly G, Tyagi M, Betsch R, Krais JJ, Taberi B, Vekariya U, Calbert M, Skorski T, Johnson N, Chen XS, and Pomerantz RT
- Subjects
- Humans, BRCA2 Protein metabolism, BRCA2 Protein genetics, BRCA2 Protein chemistry, BRCA1 Protein metabolism, BRCA1 Protein genetics, BRCA1 Protein chemistry, Piperazines pharmacology, Piperazines chemistry, Cell Line, Tumor, Phthalazines pharmacology, Phthalazines chemistry, Enzyme Inhibitors pharmacology, Enzyme Inhibitors chemistry, Models, Molecular, Adenosine Triphosphatases metabolism, Adenosine Triphosphatases antagonists & inhibitors, Protein Binding, Cryoelectron Microscopy, DNA Helicases metabolism, DNA Helicases chemistry, DNA Helicases genetics, DNA Helicases antagonists & inhibitors, DNA-Directed DNA Polymerase metabolism, DNA-Directed DNA Polymerase chemistry, DNA-Directed DNA Polymerase genetics, DNA Polymerase theta
- Abstract
DNA polymerase theta (Polθ) is a DNA helicase-polymerase protein that facilitates DNA repair and is synthetic lethal with homology-directed repair (HDR) factors. Thus, Polθ is a promising precision oncology drug-target in HDR-deficient cancers. Here, we characterize the binding and mechanism of action of a Polθ helicase (Polθ-hel) small-molecule inhibitor (AB25583) using cryo-EM. AB25583 exhibits 6 nM IC
50 against Polθ-hel, selectively kills BRCA1/2-deficient cells, and acts synergistically with olaparib in cancer cells harboring pathogenic BRCA1/2 mutations. Cryo-EM uncovers predominantly dimeric Polθ-hel:AB25583 complex structures at 3.0-3.2 Å. The structures reveal a binding-pocket deep inside the helicase central-channel, which underscores the high specificity and potency of AB25583. The cryo-EM structures in conjunction with biochemical data indicate that AB25583 inhibits the ATPase activity of Polθ-hel helicase via an allosteric mechanism. These detailed structural data and insights about AB25583 inhibition pave the way for accelerating drug development targeting Polθ-hel in HDR-deficient cancers., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
79. PARG is essential for Polθ-mediated DNA end-joining by removing repressive poly-ADP-ribose marks.
- Author
-
Vekariya U, Minakhin L, Chandramouly G, Tyagi M, Kent T, Sullivan-Reed K, Atkins J, Ralph D, Nieborowska-Skorska M, Kukuyan AM, Tang HY, Pomerantz RT, and Skorski T
- Subjects
- Humans, Poly Adenosine Diphosphate Ribose metabolism, DNA Damage, Animals, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, DNA metabolism, DNA genetics, HEK293 Cells, Poly ADP Ribosylation, Poly(ADP-ribose) Polymerases metabolism, Poly(ADP-ribose) Polymerases genetics, Carrier Proteins, Glycoside Hydrolases, Nuclear Proteins, Poly (ADP-Ribose) Polymerase-1 metabolism, Poly (ADP-Ribose) Polymerase-1 genetics, DNA Polymerase theta, DNA End-Joining Repair, DNA-Directed DNA Polymerase metabolism, DNA Breaks, Double-Stranded
- Abstract
DNA polymerase theta (Polθ)-mediated end-joining (TMEJ) repairs DNA double-strand breaks and confers resistance to genotoxic agents. How Polθ is regulated at the molecular level to exert TMEJ remains poorly characterized. We find that Polθ interacts with and is PARylated by PARP1 in a HPF1-independent manner. PARP1 recruits Polθ to the vicinity of DNA damage via PARylation dependent liquid demixing, however, PARylated Polθ cannot perform TMEJ due to its inability to bind DNA. PARG-mediated de-PARylation of Polθ reactivates its DNA binding and end-joining activities. Consistent with this, PARG is essential for TMEJ and the temporal recruitment of PARG to DNA damage corresponds with TMEJ activation and dissipation of PARP1 and PAR. In conclusion, we show a two-step spatiotemporal mechanism of TMEJ regulation. First, PARP1 PARylates Polθ and facilitates its recruitment to DNA damage sites in an inactivated state. PARG subsequently activates TMEJ by removing repressive PAR marks on Polθ., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
80. DNA damage response genes as biomarkers of therapeutic outcomes in acute myeloid leukemia patients.
- Author
-
Karami A and Skorski T
- Subjects
- Humans, Prognosis, Treatment Outcome, Male, Female, Middle Aged, DNA Repair genetics, Aged, Leukemia, Myeloid, Acute genetics, DNA Damage, Biomarkers, Tumor genetics
- Published
- 2024
- Full Text
- View/download PDF
81. 4'-Ethynyl-2'-Deoxycytidine (EdC) Preferentially Targets Lymphoma and Leukemia Subtypes by Inducing Replicative Stress.
- Author
-
Calbert ML, Chandramouly G, Adams CM, Saez-Ayala M, Kent T, Tyagi M, Ayyadevara VSSA, Wang Y, Krais JJ, Gordon J, Atkins J, Toma MM, Betzi S, Boghossian AS, Rees MG, Ronan MM, Roth JA, Goldman AR, Gorman N, Mitra R, Childers WE, Graña X, Skorski T, Johnson N, Hurtz C, Morelli X, Eischen CM, and Pomerantz RT
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Lymphoma drug therapy, Lymphoma pathology, Lymphoma metabolism, Xenograft Model Antitumor Assays, Leukemia drug therapy, Leukemia pathology, Deoxycytidine Kinase metabolism, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology
- Abstract
Anticancer nucleosides are effective against solid tumors and hematologic malignancies, but typically are prone to nucleoside metabolism resistance mechanisms. Using a nucleoside-specific multiplexed high-throughput screening approach, we discovered 4'-ethynyl-2'-deoxycytidine (EdC) as a third-generation anticancer nucleoside prodrug with preferential activity against diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukemia (ALL). EdC requires deoxycytidine kinase (DCK) phosphorylation for its activity and induces replication fork arrest and accumulation of cells in S-phase, indicating it acts as a chain terminator. A 2.1Å cocrystal structure of DCK bound to EdC and UDP reveals how the rigid 4'-alkyne of EdC fits within the active site of DCK. Remarkably, EdC was resistant to cytidine deamination and SAMHD1 metabolism mechanisms and exhibited higher potency against ALL compared with FDA-approved nelarabine. Finally, EdC was highly effective against DLBCL tumors and B-ALL in vivo. These data characterize EdC as a preclinical nucleoside prodrug candidate for DLBCL and ALL., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
82. Discovery of a small-molecule inhibitor that traps Polθ on DNA and synergizes with PARP inhibitors.
- Author
-
Fried W, Tyagi M, Minakhin L, Chandramouly G, Tredinnick T, Ramanjulu M, Auerbacher W, Calbert M, Rusanov T, Hoang T, Borisonnik N, Betsch R, Krais JJ, Wang Y, Vekariya UM, Gordon J, Morton G, Kent T, Skorski T, Johnson N, Childers W, Chen XS, and Pomerantz RT
- Subjects
- BRCA2 Protein genetics, DNA metabolism, DNA Repair, DNA-Directed DNA Polymerase metabolism, Homologous Recombination, Humans, BRCA1 Protein genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology
- Abstract
The DNA damage response (DDR) protein DNA Polymerase θ (Polθ) is synthetic lethal with homologous recombination (HR) factors and is therefore a promising drug target in BRCA1/2 mutant cancers. We discover an allosteric Polθ inhibitor (Polθi) class with 4-6 nM IC
50 that selectively kills HR-deficient cells and acts synergistically with PARP inhibitors (PARPi) in multiple genetic backgrounds. X-ray crystallography and biochemistry reveal that Polθi selectively inhibits Polθ polymerase (Polθ-pol) in the closed conformation on B-form DNA/DNA via an induced fit mechanism. In contrast, Polθi fails to inhibit Polθ-pol catalytic activity on A-form DNA/RNA in which the enzyme binds in the open configuration. Remarkably, Polθi binding to the Polθ-pol:DNA/DNA closed complex traps the polymerase on DNA for more than forty minutes which elucidates the inhibitory mechanism of action. These data reveal a unique small-molecule DNA polymerase:DNA trapping mechanism that induces synthetic lethality in HR-deficient cells and potentiates the activity of PARPi., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
83. Clonal medicine targeting DNA damage response eradicates leukemia.
- Author
-
Toma MM, Karami A, Nieborowska-Skorska M, Chirtala KN, Pepek M, Hadzijusufovic E, Stoklosa T, Valent P, and Skorski T
- Subjects
- Humans, Clone Cells, DNA Repair, DNA Damage, Leukemia genetics
- Published
- 2024
- Full Text
- View/download PDF
84. R-Loop Accumulation in Spliceosome Mutant Leukemias Confers Sensitivity to PARP1 Inhibition by Triggering Transcription-Replication Conflicts.
- Author
-
Liu ZS, Sinha S, Bannister M, Song A, Arriaga-Gomez E, McKeeken AJ, Bonner EA, Hanson BK, Sarchi M, Takashima K, Zong D, Corral VM, Nguyen E, Yoo J, Chiraphapphaiboon W, Leibson C, McMahon MC, Rai S, Swisher EM, Sachs Z, Chatla S, Stirewalt DL, Deeg HJ, Skorski T, Papapetrou EP, Walter MJ, Graubert TA, Doulatov S, Lee SC, and Nguyen HD
- Subjects
- Humans, R-Loop Structures, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, DNA Repair, RNA Splicing Factors genetics, Poly (ADP-Ribose) Polymerase-1 genetics, Spliceosomes genetics, Leukemia drug therapy, Leukemia genetics
- Abstract
RNA splicing factor (SF) gene mutations are commonly observed in patients with myeloid malignancies. Here we showed that SRSF2- and U2AF1-mutant leukemias are preferentially sensitive to PARP inhibitors (PARPi), despite being proficient in homologous recombination repair. Instead, SF-mutant leukemias exhibited R-loop accumulation that elicited an R-loop-associated PARP1 response, rendering cells dependent on PARP1 activity for survival. Consequently, PARPi induced DNA damage and cell death in SF-mutant leukemias in an R-loop-dependent manner. PARPi further increased aberrant R-loop levels, causing higher transcription-replication collisions and triggering ATR activation in SF-mutant leukemias. Ultimately, PARPi-induced DNA damage and cell death in SF-mutant leukemias could be enhanced by ATR inhibition. Finally, the level of PARP1 activity at R-loops correlated with PARPi sensitivity, suggesting that R-loop-associated PARP1 activity could be predictive of PARPi sensitivity in patients harboring SF gene mutations. This study highlights the potential of targeting different R-loop response pathways caused by spliceosome gene mutations as a therapeutic strategy for treating cancer., Significance: Spliceosome-mutant leukemias accumulate R-loops and require PARP1 to resolve transcription-replication conflicts and genomic instability, providing rationale to repurpose FDA-approved PARP inhibitors for patients carrying spliceosome gene mutations., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
85. Epigenetic balance ensures mechanistic control of MLL amplification and rearrangement.
- Author
-
Gray ZH, Chakraborty D, Duttweiler RR, Alekbaeva GD, Murphy SE, Chetal K, Ji F, Ferman BI, Honer MA, Wang Z, Myers C, Sun R, Kaniskan HÜ, Toma MM, Bondarenko EA, Santoro JN, Miranda C, Dillingham ME, Tang R, Gozani O, Jin J, Skorski T, Duy C, Lee H, Sadreyev RI, and Whetstine JR
- Subjects
- Adult, Animals, Humans, Infant, Mice, Doxorubicin pharmacology, Gene Rearrangement, Histocompatibility Antigens, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Leukemia metabolism, Lysine metabolism, Translocation, Genetic, Epigenesis, Genetic, Myeloid-Lymphoid Leukemia Protein genetics
- Abstract
MLL/KMT2A amplifications and translocations are prevalent in infant, adult, and therapy-induced leukemia. However, the molecular contributor(s) to these alterations are unclear. Here, we demonstrate that histone H3 lysine 9 mono- and di-methylation (H3K9me1/2) balance at the MLL/KMT2A locus regulates these amplifications and rearrangements. This balance is controlled by the crosstalk between lysine demethylase KDM3B and methyltransferase G9a/EHMT2. KDM3B depletion increases H3K9me1/2 levels and reduces CTCF occupancy at the MLL/KMT2A locus, in turn promoting amplification and rearrangements. Depleting CTCF is also sufficient to generate these focal alterations. Furthermore, the chemotherapy doxorubicin (Dox), which associates with therapy-induced leukemia and promotes MLL/KMT2A amplifications and rearrangements, suppresses KDM3B and CTCF protein levels. KDM3B and CTCF overexpression rescues Dox-induced MLL/KMT2A alterations. G9a inhibition in human cells or mice also suppresses MLL/KMT2A events accompanying Dox treatment. Therefore, MLL/KMT2A amplifications and rearrangements are controlled by epigenetic regulators that are tractable drug targets, which has clinical implications., Competing Interests: Declaration of interests J.R.W. has served or is serving as a consultant or advisor for Qsonica, Salarius Pharmaceuticals, Daiichi Sankyo, Inc., Vyne Therapeutics, and Lily Asia Ventures. J.R.W. also receives funding for research from Salarius Pharmaceuticals and Oryzon Genomics. O.G. is a scientific cofounder and shareholder of EpiCypher, Inc., K36 Therapeutics, Inc., and Alternative Bio, Inc. J.J. received research funds from Celgene Corporation, Levo Therapeutics, Inc., Cullgen, Inc., and Cullinan Oncology, Inc. J.J. is a cofounder and equity shareholder in Cullgen, Inc., a scientific cofounder and scientific advisory board member of Onsero Therapeutics, Inc., and a consultant for Cullgen, Inc., EpiCypher, Inc., and Accent Therapeutics, Inc. C.D. receives research funds from Janssen outside the submitted work., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
86. Simultaneous Targeting of DNA Polymerase Theta and PARP1 or RAD52 Triggers Dual Synthetic Lethality in Homologous Recombination-Deficient Leukemia Cells.
- Author
-
Sullivan-Reed K, Toma MM, Drzewiecka M, Nieborowska-Skorska M, Nejati R, Karami A, Wasik MA, Sliwinski T, and Skorski T
- Subjects
- Humans, BRCA1 Protein genetics, BRCA1 Protein metabolism, BRCA2 Protein genetics, Homologous Recombination, DNA Repair, Rad52 DNA Repair and Recombination Protein genetics, Poly (ADP-Ribose) Polymerase-1 genetics, DNA Polymerase theta, Synthetic Lethal Mutations, Leukemia genetics
- Abstract
DNA polymerase theta (Polθ, encoded by POLQ gene) plays an essential role in Polθ-mediated end-joining (TMEJ) of DNA double-strand breaks (DSB). Inhibition of Polθ is synthetic lethal in homologous recombination (HR)-deficient tumor cells. However, DSBs can be also repaired by PARP1 and RAD52-mediated mechanisms. Because leukemia cells accumulate spontaneous DSBs, we tested if simultaneous targeting of Polθ and PARP1 or RAD52 enhance the synthetic lethal effect in HR-deficient leukemia cells. Transformation potential of the oncogenes inducing BRCA1/2-deficiency (BCR-ABL1 and AML1-ETO) was severely limited in Polq-/-;Parp1-/- and Polq-/-;Rad52-/- cells when compared with single knockouts, which was associated with accumulation of DSBs. Small-molecule inhibitor of Polθ (Polθi) when combined with PARP or RAD52 inhibitors (PARPi, RAD52i) caused accumulation of DSBs and exerted increased effect against HR-deficient leukemia and myeloproliferative neoplasm cells., Implications: In conclusion, we show that PARPi or RAD52i might improve therapeutic effect of Polθi against HR-deficient leukemias., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
87. Class I HDAC Inhibition Leads to a Downregulation of FANCD2 and RAD51, and the Eradication of Glioblastoma Cells.
- Author
-
Drzewiecka M, Jaśniak D, Barszczewska-Pietraszek G, Czarny P, Kobrzycka A, Wieczorek M, Radek M, Szemraj J, Skorski T, and Śliwiński T
- Abstract
HDAC inhibitors (HDACi) hold great potential as anticancer therapies due to their ability to regulate the acetylation of both histone and non-histone proteins, which is frequently disrupted in cancer and contributes to the development and advancement of the disease. Additionally, HDACi have been shown to enhance the cytotoxic effects of DNA-damaging agents such as radiation and cisplatin. In this study, we found that histone deacetylase inhibits valproic acid (VPA), synergized with PARP1 inhibitor (PARPi), talazoparib (BMN-673), and alkylating agent, and temozolomide (TMZ) to induce DNA damage and reduce glioblastoma multiforme. At the molecular level, VPA leads to a downregulation of FANCD2 and RAD51, and the eradication of glioblastoma cells. The results of this study indicate that combining HDACi with PARPi could potentially enhance the treatment of glioblastoma, the most aggressive type of cancer that originates in the brain.
- Published
- 2023
- Full Text
- View/download PDF
88. Histone Deacetylases (HDAC) Inhibitor-Valproic Acid Sensitizes Human Melanoma Cells to Dacarbazine and PARP Inhibitor.
- Author
-
Drzewiecka M, Gajos-Michniewicz A, Hoser G, Jaśniak D, Barszczewska-Pietraszek G, Sitarek P, Czarny P, Piekarski J, Radek M, Czyż M, Skorski T, and Śliwiński T
- Subjects
- Humans, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Valproic Acid pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Dacarbazine therapeutic use, Histone Deacetylases genetics, Histone Deacetylases metabolism, DNA, Alkylating Agents therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology
- Abstract
The inhibition of histone deacetylases (HDACs) holds promise as a potential anti-cancer therapy as histone and non-histone protein acetylation is frequently disrupted in cancer, leading to cancer initiation and progression. Additionally, the use of a histone deacetylase inhibitor (HDACi) such as the class I HDAC inhibitor-valproic acid (VPA) has been shown to enhance the effectiveness of DNA-damaging factors, such as cisplatin or radiation. In this study, we found that the use of VPA in combination with talazoparib (BMN-673-PARP1 inhibitor-PARPi) and/or Dacarbazine (DTIC-alkylating agent) resulted in an increased rate of DNA double strand breaks (DSBs) and reduced survival (while not affecting primary melanocytes) and the proliferation of melanoma cells. Furthermore, the pharmacological inhibition of class I HDACs sensitizes melanoma cells to apoptosis following exposure to DTIC and BMN-673. In addition, the inhibition of HDACs causes the sensitization of melanoma cells to DTIV and BMN-673 in melanoma xenografts in vivo. At the mRNA and protein level, the histone deacetylase inhibitor downregulated RAD51 and FANCD2. This study aims to demonstrate that combining an HDACi, alkylating agent and PARPi could potentially enhance the treatment of melanoma, which is commonly recognized as being among the most aggressive malignant tumors. The findings presented here point to a scenario in which HDACs, via enhancing the HR-dependent repair of DSBs created during the processing of DNA lesions, are essential nodes in the resistance of malignant melanoma cells to methylating agent-based therapies.
- Published
- 2023
- Full Text
- View/download PDF
89. DNA polymerase θ protects leukemia cells from metabolically induced DNA damage.
- Author
-
Vekariya U, Toma M, Nieborowska-Skorska M, Le BV, Caron MC, Kukuyan AM, Sullivan-Reed K, Podszywalow-Bartnicka P, Chitrala KN, Atkins J, Drzewiecka M, Feng W, Chan J, Chatla S, Golovine K, Jelinek J, Sliwinski T, Ghosh J, Matlawska-Wasowska K, Chandramouly G, Nejati R, Wasik M, Sykes SM, Piwocka K, Hadzijusufovic E, Valent P, Pomerantz RT, Morton G, Childers W, Zhao H, Paietta EM, Levine RL, Tallman MS, Fernandez HF, Litzow MR, Gupta GP, Masson JY, and Skorski T
- Subjects
- Animals, Mice, BRCA2 Protein, DNA metabolism, DNA Polymerase theta, BRCA1 Protein, DNA Damage, Leukemia enzymology, Leukemia genetics
- Abstract
Leukemia cells accumulate DNA damage, but altered DNA repair mechanisms protect them from apoptosis. We showed here that formaldehyde generated by serine/1-carbon cycle metabolism contributed to the accumulation of toxic DNA-protein crosslinks (DPCs) in leukemia cells, especially in driver clones harboring oncogenic tyrosine kinases (OTKs: FLT3(internal tandem duplication [ITD]), JAK2(V617F), BCR-ABL1). To counteract this effect, OTKs enhanced the expression of DNA polymerase theta (POLθ) via ERK1/2 serine/threonine kinase-dependent inhibition of c-CBL E3 ligase-mediated ubiquitination of POLθ and its proteasomal degradation. Overexpression of POLθ in OTK-positive cells resulted in the efficient repair of DPC-containing DNA double-strand breaks by POLθ-mediated end-joining. The transforming activities of OTKs and other leukemia-inducing oncogenes, especially of those causing the inhibition of BRCA1/2-mediated homologous recombination with and without concomitant inhibition of DNA-PK-dependent nonhomologous end-joining, was abrogated in Polq-/- murine bone marrow cells. Genetic and pharmacological targeting of POLθ polymerase and helicase activities revealed that both activities are promising targets in leukemia cells. Moreover, OTK inhibitors or DPC-inducing drug etoposide enhanced the antileukemia effect of POLθ inhibitor in vitro and in vivo. In conclusion, we demonstrated that POLθ plays an essential role in protecting leukemia cells from metabolically induced toxic DNA lesions triggered by formaldehyde, and it can be targeted to achieve a therapeutic effect., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
90. Haploinsufficiency of ZNF251 causes DNA-PKcs-dependent resistance to PARP inhibitors in BRCA1-mutated cancer cells.
- Author
-
Li H, Chatla S, Liu X, Vekariya U, Kim D, Walt M, Lian Z, Morton G, Feng Z, Yang D, Liu H, Reed K, Childers W, Yu X, Madzo J, Chitrala KN, Skorski T, and Huang J
- Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors represent a promising new class of agents that have demonstrated efficacy in treating various cancers, particularly those that carry BRCA1/2 mutations. The cancer associated BRCA1/2 mutations disrupt DNA double strand break (DSB) repair by homologous recombination (HR). PARP inhibitors (PARPis) have been applied to trigger synthetic lethality in BRCA1/2 -mutated cancer cells by promoting the accumulation of toxic DSBs. Unfortunately, resistance to PARPis is common and can occur through multiple mechanisms, including the restoration of HR and/or the stabilization of replication forks. To gain a better understanding of the mechanisms underlying PARPi resistance, we conducted an unbiased CRISPR-pooled genome-wide library screen to identify new genes whose deficiency confers resistance to the PARPi olaparib. Our study revealed that ZNF251, a transcription factor, is a novel gene whose haploinsufficiency confers PARPi resistance in multiple breast and ovarian cancer lines harboring BRCA1 mutations. Mechanistically, we discovered that ZNF251 haploinsufficiency leads to constitutive stimulation of DNA-PKcs-dependent non-homologous end joining (NHEJ) repair of DSBs and DNA-PKcs-mediated fork protection in BRCA1 -mutated cancer cells (BRCA1mut + ZNF251 KD). Moreover, we demonstrated that DNA-PKcs inhibitors can restore PARPi sensitivity in BRCA1mut + ZNF251 KD cells ex vivo and in vivo . Our findings provide important insights into the mechanisms underlying PARPi resistance and highlight the unexpected role of DNA-PKcs in this phenomenon., Competing Interests: Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed by any of the authors.
- Published
- 2023
- Full Text
- View/download PDF
91. TIAR and FMRP shape pro-survival nascent proteome of leukemia cells in the bone marrow microenvironment.
- Author
-
Wolczyk M, Serwa R, Kominek A, Klejman A, Milek J, Chwałek M, Turos-Korgul L, Charzyńska A, Dabrowski M, Dziembowska M, Skorski T, Piwocka K, and Podszywalow-Bartnicka P
- Abstract
Chronic myeloid leukemia (CML) cells circulate between blood and bone marrow niche, representing different microenvironments. We studied the role of the two RNA-binding proteins, T-cell-restricted intracellular antigen (TIAR), and the fragile X mental retardation protein (FMRP) in the regulation of protein translation in CML cells residing in settings mimicking peripheral blood microenvironment (PBM) and bone marrow microenvironment (BMM). The outcomes showed how conditions shaped the translation process through TIAR and FMRP activity, considering its relevance in therapy resistance. The QuaNCAT mass-spectrometric approach revealed that TIAR and FMRP have a discrete modulatory effect on protein synthesis and thus affect distinct aspects of leukemic cells functioning in the hypoxic niche. In the BMM setup, FMRP impacted metabolic adaptation of cells and TIAR substantially supported the resistance of CML cells to translation inhibition by homoharringtonine. Overall, our results demonstrated that targeting post-transcriptional control should be considered when designing anti-leukemia therapeutic solutions., Competing Interests: The authors declare no competing interests., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
92. ABL1 kinase as a tumor suppressor in AML1-ETO and NUP98-PMX1 leukemias.
- Author
-
Golovine K, Abalakov G, Lian Z, Chatla S, Karami A, Chitrala KN, Madzo J, Nieborowska-Skorska M, Huang J, and Skorski T
- Subjects
- Animals, Humans, Mice, Core Binding Factor Alpha 2 Subunit genetics, Nuclear Pore Complex Proteins genetics, Nuclear Pore Complex Proteins metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, RUNX1 Translocation Partner 1 Protein genetics, Proto-Oncogene Proteins c-abl metabolism, Leukemia, Phosphatidylinositol 3-Kinases metabolism
- Abstract
Deletion of ABL1 was detected in a cohort of hematologic malignancies carrying AML1-ETO and NUP98 fusion proteins. Abl1-/- murine hematopoietic cells transduced with AML1-ETO and NUP98-PMX1 gained proliferation advantage when compared to Abl1 + /+ counterparts. Conversely, overexpression and pharmacological stimulation of ABL1 kinase resulted in reduced proliferation. To pinpoint mechanisms facilitating the transformation of ABL1-deficient cells, Abl1 was knocked down in 32Dcl3-Abl1ko cells by CRISPR/Cas9 followed by the challenge of growth factor withdrawal. 32Dcl3-Abl1ko cells but not 32Dcl3-Abl1wt cells generated growth factor-independent clones. RNA-seq implicated PI3K signaling as one of the dominant mechanisms contributing to growth factor independence in 32Dcl3-Abl1ko cells. PI3K inhibitor buparlisib exerted selective activity against Lin-cKit+ NUP98-PMX1;Abl1-/- cells when compared to the Abl1 + /+ counterparts. Since the role of ABL1 in DNA damage response (DDR) is well established, we also tested the inhibitors of ATM (ATMi), ATR (ATRi) and DNA-PKcs (DNA-PKi). AML1-ETO;Abl1-/- and NUP98-PMX1;Abl1-/- cells were hypersensitive to DNA-PKi and ATRi, respectively, when compared to Abl1 + /+ counterparts. Moreover, ABL1 kinase inhibitor enhanced the sensitivity to PI3K, DNA-PKcs and ATR inhibitors. In conclusion, we showed that ABL1 kinase plays a tumor suppressor role in hematological malignancies induced by AML1-ETO and NUP98-PMX1 and modulates the response to PI3K and/or DDR inhibitors., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
93. Polθ Inhibition: An Anticancer Therapy for HR-Deficient Tumours.
- Author
-
Barszczewska-Pietraszek G, Drzewiecka M, Czarny P, Skorski T, and Śliwiński T
- Subjects
- Humans, DNA genetics, DNA Breaks, Double-Stranded, DNA End-Joining Repair, Homologous Recombination, DNA Polymerase theta, DNA Repair, Neoplasms drug therapy, Neoplasms genetics, Nucleic Acid Synthesis Inhibitors pharmacology
- Abstract
DNA polymerase theta (Polθ)-mediated end joining (TMEJ) is, along with homologous recombination (HR) and non-homologous end-joining (NHEJ), one of the most important mechanisms repairing potentially lethal DNA double-strand breaks (DSBs). Polθ is becoming a new target in cancer research because it demonstrates numerous synthetically lethal interactions with other DNA repair mechanisms, e.g., those involving PARP1, BRCA1/2, DNA-PK, ATR. Inhibition of Polθ could be achieved with different methods, such as RNA interference (RNAi), CRISPR/Cas9 technology, or using small molecule inhibitors. In the context of this topic, RNAi and CRISPR/Cas9 are still more often applied in the research itself rather than clinical usage, different than small molecule inhibitors. Several Polθ inhibitors have been already generated, and two of them, novobiocin (NVB) and ART812 derivative, are being tested in clinical trials against HR-deficient tumors. In this review, we describe the significance of Polθ and the Polθ-mediated TMEJ pathway. In addition, we summarize the current state of knowledge about Polθ inhibitors and emphasize the promising role of Polθ as a therapeutic target.
- Published
- 2022
- Full Text
- View/download PDF
94. Pre-Existing and Acquired Resistance to PARP Inhibitor-Induced Synthetic Lethality.
- Author
-
Le BV, Podszywałow-Bartnicka P, Piwocka K, and Skorski T
- Abstract
The advanced development of synthetic lethality has opened the doors for specific anti-cancer medications of personalized medicine and efficient therapies against cancers. One of the most popular approaches being investigated is targeting DNA repair pathways as the implementation of the PARP inhibitor (PARPi) into individual or combinational therapeutic schemes. Such treatment has been effectively employed against homologous recombination-defective solid tumors as well as hematopoietic malignancies. However, the resistance to PARPi has been observed in both preclinical research and clinical treatment. Therefore, elucidating the mechanisms responsible for the resistance to PARPi is pivotal for the further success of this intervention. Apart from mechanisms of acquired resistance, the bone marrow microenvironment provides a pre-existing mechanism to induce the inefficiency of PARPi in leukemic cells. Here, we describe the pre-existing and acquired mechanisms of the resistance to PARPi-induced synthetic lethality. We also discuss the potential rationales for developing effective therapies to prevent/repress the PARPi resistance in cancer cells.
- Published
- 2022
- Full Text
- View/download PDF
95. Perspective: Pivotal translational hematology and therapeutic insights in chronic myeloid hematopoietic stem cell malignancies.
- Author
-
Mughal TI, Pemmaraju N, Bejar R, Gale RP, Bose P, Kiladjian JJ, Prchal J, Royston D, Pollyea D, Valent P, Brümmendorf TH, Skorski T, Patnaik M, Santini V, Fenaux P, Kucine N, Verstovsek S, Mesa R, Barbui T, Saglio G, and Van Etten RA
- Subjects
- Hematopoietic Stem Cells, Humans, COVID-19, Graft vs Host Disease, Hematology, Myeloproliferative Disorders drug therapy
- Abstract
Despite much of the past 2 years being engulfed by the devastating consequences of the SAR-CoV-2 pandemic, significant progress, even breathtaking, occurred in the field of chronic myeloid malignancies. Some of this was show-cased at the 15th Post-American Society of Hematology (ASH) and the 25th John Goldman workshops on myeloproliferative neoplasms (MPN) held on 9th-10th December 2020 and 7th-10th October 2021, respectively. The inaugural Post-ASH MPN workshop was set out in 2006 by John Goldman (deceased) and Tariq Mughal to answer emerging translational hematology and therapeutics of patients with these malignancies. Rather than present a resume of the discussions, this perspective focuses on some of the pivotal translational hematology and therapeutic insights in these diseases., (© 2022 John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
96. Synthetic Lethality Targeting Polθ.
- Author
-
Drzewiecka M, Barszczewska-Pietraszek G, Czarny P, Skorski T, and Śliwiński T
- Subjects
- DNA Breaks, Double-Stranded, DNA Repair genetics, Humans, Recombinational DNA Repair, Neoplasms drug therapy, Neoplasms genetics, Synthetic Lethal Mutations genetics
- Abstract
Research studies regarding synthetic lethality (SL) in human cells are primarily motivated by the potential of this phenomenon to be an effective, but at the same time, safe to the patient's anti-cancer chemotherapy. Among the factors that are targets for the induction of the synthetic lethality effect, those involved in DNA repair seem to be the most relevant. Specifically, when mutation in one of the canonical DNA double-strand break (DSB) repair pathways occurs, which is a frequent event in cancer cells, the alternative pathways may be a promising target for the elimination of abnormal cells. Currently, inhibiting RAD52 and/or PARP1 in the tumor cells that are deficient in the canonical repair pathways has been the potential target for inducing the effect of synthetic lethality. Unfortunately, the development of resistance to commonly used PARP1 inhibitors (PARPi) represents the greatest obstacle to working out a successful treatment protocol. DNA polymerase theta (Polθ), encoded by the POLQ gene, plays a key role in an alternative DSB repair pathway-theta-mediated end joining (TMEJ). Thus, it is a promising target in the treatment of tumors harboring deficiencies in homologous recombination repair (HRR), where its inhibition can induce SL. In this review, the authors discuss the current state of knowledge on Polθ as a potential target for synthetic lethality-based anticancer therapies.
- Published
- 2022
- Full Text
- View/download PDF
97. 4-1BBL-containing leukemic extracellular vesicles promote immunosuppressive effector regulatory T cells.
- Author
-
Swatler J, Turos-Korgul L, Brewinska-Olchowik M, De Biasi S, Dudka W, Le BV, Kominek A, Cyranowski S, Pilanc P, Mohammadi E, Cysewski D, Kozlowska E, Grabowska-Pyrzewicz W, Wojda U, Basak G, Mieczkowski J, Skorski T, Cossarizza A, and Piwocka K
- Subjects
- Animals, Immunosuppressive Agents therapeutic use, Ki-1 Antigen metabolism, Mice, Receptors, Tumor Necrosis Factor, Type II genetics, Receptors, Tumor Necrosis Factor, Type II metabolism, T-Lymphocytes, Regulatory, 4-1BB Ligand immunology, Extracellular Vesicles metabolism, Leukemia, Myeloid, Acute drug therapy
- Abstract
Chronic and acute myeloid leukemia evade immune system surveillance and induce immunosuppression by expanding proleukemic Foxp3+ regulatory T cells (Tregs). High levels of immunosuppressive Tregs predict inferior response to chemotherapy, leukemia relapse, and shorter survival. However, mechanisms that promote Tregs in myeloid leukemias remain largely unexplored. Here, we identify leukemic extracellular vesicles (EVs) as drivers of effector proleukemic Tregs. Using mouse model of leukemia-like disease, we found that Rab27a-dependent secretion of leukemic EVs promoted leukemia engraftment, which was associated with higher abundance of activated, immunosuppressive Tregs. Leukemic EVs attenuated mTOR-S6 and activated STAT5 signaling, as well as evoked significant transcriptomic changes in Tregs. We further identified specific effector signature of Tregs promoted by leukemic EVs. Leukemic EVs-driven Tregs were characterized by elevated expression of effector/tumor Treg markers CD39, CCR8, CD30, TNFR2, CCR4, TIGIT, and IL21R and included 2 distinct effector Treg (eTreg) subsets: CD30+CCR8hiTNFR2hi eTreg1 and CD39+TIGIThi eTreg2. Finally, we showed that costimulatory ligand 4-1BBL/CD137L, shuttled by leukemic EVs, promoted suppressive activity and effector phenotype of Tregs by regulating expression of receptors such as CD30 and TNFR2. Collectively, our work highlights the role of leukemic extracellular vesicles in stimulation of immunosuppressive Tregs and leukemia growth. We postulate that targeting of Rab27a-dependent secretion of leukemic EVs may be a viable therapeutic approach in myeloid neoplasms., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
98. TET2 and DNMT3A Mutations Exert Divergent Effects on DNA Repair and Sensitivity of Leukemia Cells to PARP Inhibitors.
- Author
-
Maifrede S, Le BV, Nieborowska-Skorska M, Golovine K, Sullivan-Reed K, Dunuwille WMB, Nacson J, Hulse M, Keith K, Madzo J, Caruso LB, Gazze Z, Lian Z, Padella A, Chitrala KN, Bartholdy BA, Matlawska-Wasowska K, Di Marcantonio D, Simonetti G, Greiner G, Sykes SM, Valent P, Paietta EM, Tallman MS, Fernandez HF, Litzow MR, Minden MD, Huang J, Martinelli G, Vassiliou GS, Tempera I, Piwocka K, Johnson N, Challen GA, and Skorski T
- Subjects
- Animals, CRISPR-Cas Systems, Cell Line, Tumor, Disease Models, Animal, Dose-Response Relationship, Drug, Gene Knockdown Techniques, Genotype, Humans, Leukemia, Mice, Mice, Transgenic, Models, Biological, Neoplastic Stem Cells, Xenograft Model Antitumor Assays, DNA Methyltransferase 3A genetics, DNA Repair, DNA-Binding Proteins genetics, Dioxygenases genetics, Drug Resistance, Neoplasm genetics, Mutation, Poly(ADP-ribose) Polymerase Inhibitors pharmacology
- Abstract
Somatic variants in TET2 and DNMT3A are founding mutations in hematological malignancies that affect the epigenetic regulation of DNA methylation. Mutations in both genes often co-occur with activating mutations in genes encoding oncogenic tyrosine kinases such as FLT3
ITD , BCR-ABL1, JAK2V617F , and MPLW515L , or with mutations affecting related signaling pathways such as NRASG12D and CALRdel52 . Here, we show that TET2 and DNMT3A mutations exert divergent roles in regulating DNA repair activities in leukemia cells expressing these oncogenes. Malignant TET2-deficient cells displayed downregulation of BRCA1 and LIG4, resulting in reduced activity of BRCA1/2-mediated homologous recombination (HR) and DNA-PK-mediated non-homologous end-joining (D-NHEJ), respectively. TET2-deficient cells relied on PARP1-mediated alternative NHEJ (Alt-NHEJ) for protection from the toxic effects of spontaneous and drug-induced DNA double-strand breaks. Conversely, DNMT3A-deficient cells favored HR/D-NHEJ owing to downregulation of PARP1 and reduction of Alt-NHEJ. Consequently, malignant TET2-deficient cells were sensitive to PARP inhibitor (PARPi) treatment in vitro and in vivo , whereas DNMT3A-deficient cells were resistant. Disruption of TET2 dioxygenase activity or TET2-Wilms' tumor 1 (WT1)-binding ability was responsible for DNA repair defects and sensitivity to PARPi associated with TET2 deficiency. Moreover, mutation or deletion of WT1 mimicked the effect of TET2 mutation on DSB repair activity and sensitivity to PARPi. Collectively, these findings reveal that TET2 and WT1 mutations may serve as biomarkers of synthetic lethality triggered by PARPi, which should be explored therapeutically. SIGNIFICANCE: TET2 and DNMT3A mutations affect distinct DNA repair mechanisms and govern the differential sensitivities of oncogenic tyrosine kinase-positive malignant hematopoietic cells to PARP inhibitors., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
99. ATF3 coordinates serine and nucleotide metabolism to drive cell cycle progression in acute myeloid leukemia.
- Author
-
Di Marcantonio D, Martinez E, Kanefsky JS, Huhn JM, Gabbasov R, Gupta A, Krais JJ, Peri S, Tan Y, Skorski T, Dorrance A, Garzon R, Goldman AR, Tang HY, Johnson N, and Sykes SM
- Subjects
- Activating Transcription Factor 3 genetics, Cell Line, Tumor, Humans, Leukemia, Myeloid, Acute genetics, Neoplasm Proteins genetics, Nucleotides genetics, Serine genetics, Activating Transcription Factor 3 metabolism, Cell Cycle, Leukemia, Myeloid, Acute metabolism, Neoplasm Proteins metabolism, Nucleotides metabolism, Serine metabolism
- Abstract
Metabolic reprogramming is a common feature of many human cancers, including acute myeloid leukemia (AML). However, the upstream regulators that promote AML metabolic reprogramming and the benefits conferred to leukemia cells by these metabolic changes remain largely unknown. We report that the transcription factor ATF3 coordinates serine and nucleotide metabolism to maintain cell cycling, survival, and the differentiation blockade in AML. Analysis of mouse and human AML models demonstrate that ATF3 directly activates the transcription of genes encoding key enzymatic regulators of serine synthesis, one-carbon metabolism, and de novo purine and pyrimidine synthesis. Total steady-state polar metabolite and heavy isotope tracing analyses show that ATF3 inhibition reduces de novo serine synthesis, impedes the incorporation of serine-derived carbons into newly synthesized purines, and disrupts pyrimidine metabolism. Importantly, exogenous nucleotide supplementation mitigates the anti-leukemia effects of ATF3 inhibition. Together, these findings reveal the dependence of AML on ATF3-regulated serine and nucleotide metabolism., Competing Interests: Declaration of interests S.P. is currently an employee of Merck Research Laboratories and R. Gabbasov is currently an employee of Carisma Therapeutics. All other authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
100. Relationship between Oxidative Stress and Imatinib Resistance in Model Chronic Myeloid Leukemia Cells.
- Author
-
Głowacki S, Synowiec E, Szwed M, Toma M, Skorski T, and Śliwiński T
- Subjects
- Animals, Catalase metabolism, Cell Line, Tumor, DNA Damage, Fusion Proteins, bcr-abl genetics, Glutathione metabolism, Glutathione Peroxidase metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Membrane Potential, Mitochondrial, Mice, Antineoplastic Agents toxicity, Drug Resistance, Neoplasm, Imatinib Mesylate toxicity, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Oxidative Stress
- Abstract
Chronic myeloid leukemia (CML) develops due to the presence of the BCR-ABL1 protein, a target of tyrosine kinase inhibitors (TKIs), such as imatinib (IM), used in a CML therapy. CML eradication is a challenge due to developing resistance to TKIs. BCR-ABL1 induces endogenous oxidative stress leading to genomic instability and development of TKI resistance. Model CML cells susceptible or resistant to IM, as well as wild-type, non-cancer cells without the BCR-ABL1 protein were treated with IM, hydrogen peroxide (H
2 O2 ) as a model trigger of external oxidative stress, or with IM+H2 O2 . Accumulation of reactive oxygen species (ROS), DNA damage, activity of selected antioxidant enzymes and glutathione (GSH), and mitochondrial potential (MMP) were assessed. We observed increase in ROS accumulation in BCR-ABL1 positive cells and distinct levels of ROS accumulation in IM-susceptible cells when compared to IM-resistant ones, as well as increased DNA damage caused by IM action in sensitive cells. Depletion of GSH levels and a decreased activity of glutathione peroxidase (GPx) in the presence of IM was higher in the cells susceptible to IM. IM-resistant cells showed an increase of catalase activity and a depletion of MMP. BCR-ABL1 kinase alters ROS metabolism, and IM resistance is accompanied by the changes in activity of GPx, catalase, and alterations in MMP.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.