scispace - formally typeset
Search or ask a question
Author

Kartika Venugopal

Bio: Kartika Venugopal is an academic researcher from University of Florida. The author has contributed to research in topics: DNA damage & Cytarabine. The author has an hindex of 1, co-authored 5 publications receiving 7 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: Recent advances in understanding of the biochemical and structural consequences of DNMT3A mutations on DNA methylation catalysis and binding interactions are discussed and their effects on epigenetic patterns and gene expression changes implicated in the pathogenesis of hematologic malignancies are summarized.
Abstract: In the last decade, large-scale genomic studies in patients with hematologic malignancies identified recurrent somatic alterations in epigenetic modifier genes. Among these, the de novo DNA methyltransferase DNMT3A has emerged as one of the most frequently mutated genes in adult myeloid as well as lymphoid malignancies and in clonal hematopoiesis. In this review, we discuss recent advances in our understanding of the biochemical and structural consequences of DNMT3A mutations on DNA methylation catalysis and binding interactions and summarize their effects on epigenetic patterns and gene expression changes implicated in the pathogenesis of hematologic malignancies. We then review the role played by mutant DNMT3A in clonal hematopoiesis, accompanied by its effect on immune cell function and inflammatory responses. Finally, we discuss how this knowledge informs therapeutic approaches for hematologic malignancies with mutant DNMT3A.

11 citations

Journal ArticleDOI
TL;DR: It is reported that malignant pleural mesothelioma cells robustly express STING and are responsive to STING agonist treatment ex vivo, and the fundamental importance of using human tumor samples to assess innate and cellular immune therapies is revealed.
Abstract: Activation of the stimulator of interferon genes (STING) pathway promotes antitumor immunity but STING agonists have yet to achieve clinical success. Increased understanding of the mechanism of action of STING agonists in human tumors is key to developing therapeutic combinations that activate effective innate antitumor immunity. Here, we report that malignant pleural mesothelioma cells robustly express STING and are responsive to STING agonist treatment ex vivo. Using dynamic single-cell RNA sequencing of explants treated with a STING agonist we observed CXCR3 chemokine activation primarily in tumor cells and cancer-associated fibroblasts, as well as T-cell cytotoxicity. In contrast, primary NK cells resisted STING agonist-induced cytotoxicity. STING agonists enhanced migration and killing of NK cells and mesothelin-targeted chimeric antigen receptor (CAR)-NK cells, improving therapeutic activity in patient-derived organotypic tumor spheroids. These studies reveal the fundamental importance of using human tumor samples to assess innate and cellular immune therapies. By functionally profiling mesothelioma tumor explants with elevated STING expression in tumor cells, we uncovered distinct consequences of STING agonist treatment in humans that support testing combining STING agonists with NK and CAR-NK cell therapies.

6 citations

Journal ArticleDOI
TL;DR: Venugopal et al. as discussed by the authors discussed PARP inhibition as a rational strategy to increase cytarabine efficacy in cells without DNMT3A mutations, while considering the implications of PARP inhibitor resistance for promoting clonal hematopoiesis.

2 citations

Posted ContentDOI
28 May 2021-bioRxiv
TL;DR: In this article, pharmacologically-induced replication fork stalling creates a therapeutic vulnerability in cells with DNMT3A(R882) mutations, which is associated with resistance to standard chemotherapy, disease relapse, and poor prognosis.
Abstract: Mutations in the DNA methyltransferase 3A (DNMT3A) gene are recurrent in de novo acute myeloid leukemia (AML) and are associated with resistance to standard chemotherapy, disease relapse, and poor prognosis, especially in advanced-age patients. Previous gene expression studies in cells with DNMT3A mutations identified deregulation of cell cycle-related signatures implicated in DNA damage response and replication fork integrity, suggesting sensitivity to replication stress. Here we tested whether pharmacologically-induced replication fork stalling creates a therapeutic vulnerability in cells with DNMT3A(R882) mutations. We observed increased sensitivity to nucleoside analogs such as cytarabine in multiple cellular systems expressing mutant DNMT3A, ectopically or endogenously, in vitro and in vivo. Analysis of DNA damage signaling in response to cytarabine revealed persistent intra-S phase checkpoint activation, accompanied by accumulation of DNA damage in the DNMT3A(R882) overexpressing cells, which was only partially resolved after drug removal and carried through mitosis, resulting in micronucleation. Pulse-chase double-labeling experiments with EdU and BrdU after cytarabine wash-out demonstrated that cells with DNMT3A(mut) were able to restart replication but showed a higher rate of fork collapse. Gene expression profiling by RNA-seq identified deregulation of pathways associated with cell cycle progression and p53 activation, as well as metabolism and chromatin. Together, our studies show that cells with DNMT3A mutations have a defect in recovery from replication fork arrest and subsequent accumulation of unresolved DNA damage, which may have therapeutic tractability. These results demonstrate that, in addition to its role in epigenetic control, DNMT3A contributes to preserving genome integrity during DNA replication.

2 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: In this article , the authors discuss how environmental contexts associated with CHIP, such as old age, infections, chemotherapy, or cigarette smoking, alter tissue microenvironments to facilitate the selection and expansion of specific CHIP mutant clones.

23 citations

Journal ArticleDOI
TL;DR: Recent advances in understanding of the biochemical and structural consequences of DNMT3A mutations on DNA methylation catalysis and binding interactions are discussed and their effects on epigenetic patterns and gene expression changes implicated in the pathogenesis of hematologic malignancies are summarized.
Abstract: In the last decade, large-scale genomic studies in patients with hematologic malignancies identified recurrent somatic alterations in epigenetic modifier genes. Among these, the de novo DNA methyltransferase DNMT3A has emerged as one of the most frequently mutated genes in adult myeloid as well as lymphoid malignancies and in clonal hematopoiesis. In this review, we discuss recent advances in our understanding of the biochemical and structural consequences of DNMT3A mutations on DNA methylation catalysis and binding interactions and summarize their effects on epigenetic patterns and gene expression changes implicated in the pathogenesis of hematologic malignancies. We then review the role played by mutant DNMT3A in clonal hematopoiesis, accompanied by its effect on immune cell function and inflammatory responses. Finally, we discuss how this knowledge informs therapeutic approaches for hematologic malignancies with mutant DNMT3A.

11 citations

Journal ArticleDOI
TL;DR: In this article , the metabolic and cellular barriers that inhibit NK cells in solid neoplasms are reviewed and potential strategies to circumvent such obstacles towards superior therapeutic activity are discussed, as well as potential strategies for circumventing such obstacles.
Abstract: Abstract Natural killer (NK) cells, which are innate lymphocytes endowed with potent cytotoxic activity, have recently attracted attention as potential anticancer therapeutics. While NK cells mediate encouraging responses in patients with leukemia, the therapeutic effects of NK cell infusion in patients with solid tumors are limited. Preclinical and clinical data suggest that the efficacy of NK cell infusion against solid malignancies is hampered by several factors including inadequate tumor infiltration and persistence/activation in the tumor microenvironment (TME). A number of metabolic features of the TME including hypoxia as well as elevated levels of adenosine, reactive oxygen species, and prostaglandins negatively affect NK cell activity. Moreover, cancer-associated fibroblasts, tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells actively suppress NK cell-dependent anticancer immunity. Here, we review the metabolic and cellular barriers that inhibit NK cells in solid neoplasms as we discuss potential strategies to circumvent such obstacles towards superior therapeutic activity.

11 citations

Journal ArticleDOI
TL;DR: In this paper , transient MPS1 inhibition (MPS1i) was shown to re-engage this pathway in KL cells via micronuclei generation, and a single course of decitabine treatment followed by pulse MPS 1i therapy restores T cell infiltration in vivo, enhances anti-PD 1 efficacy, and results in a durable response without evidence of significant toxicity.

11 citations

Journal ArticleDOI
TL;DR: In this article, a review article summarises the most remarkable findings demonstrating a contribution of epigenetic factors as proper "drivers" of tumorigenesis, and also try to pose attention on the relevance of the epigenetic mechanisms as downstream consequences of genes versus environment interaction.

8 citations