Resistance to platinum-based cancer drugs: a special focus on epigenetic mechanisms

2021 ◽  
Author(s):  
Yuselin Mora ◽  
María Elena Reyes ◽  
Louise Zanella ◽  
Bárbara Mora ◽  
Kurt Buchegger ◽  
...  

Chemoresistance is a significant clinical challenge, limiting the drug response in cancer. Several mechanisms associated with drug resistance have been characterized, and the role of epigenetics in generating resistance to platinum-based drugs has been clarified. Epigenetic mechanisms such as DNA methylation, histone modification, long noncoding RNA, and microRNA affect the expression of genes implicated in absorption, distribution, metabolism and excretion (ADME) of drugs, and other non-ADME genes that encode enzymes involved in the processes of cell proliferation, DNA repair, apoptosis and signal transduction key in the development of chemoresistance in cancer, specifically in platinum-based drugs. This review summarizes current discoveries in epigenetic regulation implicated in platinum drug resistance in cancer and the main clinical trials based on epigenetic therapy, evaluating their potential synergy with platinum-based drugs.

2021 ◽  
Vol 22 (5) ◽  
pp. 2529
Author(s):  
Amin Javadifar ◽  
Sahar Rastgoo ◽  
Maciej Banach ◽  
Tannaz Jamialahmadi ◽  
Thomas P. Johnston ◽  
...  

Atherosclerosis is a major cause of human cardiovascular disease, which is the leading cause of mortality around the world. Various physiological and pathological processes are involved, including chronic inflammation, dysregulation of lipid metabolism, development of an environment characterized by oxidative stress and improper immune responses. Accordingly, the expansion of novel targets for the treatment of atherosclerosis is necessary. In this study, we focus on the role of foam cells in the development of atherosclerosis. The specific therapeutic goals associated with each stage in the formation of foam cells and the development of atherosclerosis will be considered. Processing and metabolism of cholesterol in the macrophage is one of the main steps in foam cell formation. Cholesterol processing involves lipid uptake, cholesterol esterification and cholesterol efflux, which ultimately leads to cholesterol equilibrium in the macrophage. Recently, many preclinical studies have appeared concerning the role of non-encoding RNAs in the formation of atherosclerotic lesions. Non-encoding RNAs, especially microRNAs, are considered regulators of lipid metabolism by affecting the expression of genes involved in the uptake (e.g., CD36 and LOX1) esterification (ACAT1) and efflux (ABCA1, ABCG1) of cholesterol. They are also able to regulate inflammatory pathways, produce cytokines and mediate foam cell apoptosis. We have reviewed important preclinical evidence of their therapeutic targeting in atherosclerosis, with a special focus on foam cell formation.


Author(s):  
Ashutosh Singh ◽  
Ashutosh Kumar Singh ◽  
Rajanish Giri ◽  
Dhruv Kumar ◽  
Rohit Sharma ◽  
...  

MicroRNAs (miRNAs), a class of small noncoding RNA, posttranscriptionally regulate the expression of genes. Aberrant expression of miRNA is reported in various types of cancer. Since the first report of oncomiR-21 involvement in the glioma, its upregulation was reported in multiple cancers and was allied with high oncogenic property. In addition to the downregulation of tumor suppressor genes, the miR-21 is also associated with cancer resistance to various chemotherapy. The recent research is appraising miR-21 as a promising cancer target and biomarker for early cancer detection. In this review, we briefly explain the biogenesis and regulation of miR-21 in cancer cells. Additionally, the review features the assorted genes/pathways regulated by the miR-21 in various cancer and cancer stem cells.


Cancers ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 784 ◽  
Author(s):  
Durga Khandekar ◽  
Venkataswarup Tiriveedhi

Bromodomain and extraterminal domain (BET) proteins have evolved as key multifunctional super-regulators that control gene expression. These proteins have been shown to upregulate transcriptional machinery leading to over expression of genes involved in cell proliferation and carcinogenesis. Based on favorable preclinical evidence of BET inhibitors in various cancer models; currently, 26 clinical trials are underway in various stages of study on various hematological and solid organ cancers. Unfortunately, preliminary evidence for these clinical studies does not support the application of BET inhibitors as monotherapy in cancer treatment. Furthermore, the combinatorial efficiency of BET inhibitors with other chemo-and immunotherapeutic agents remain elusive. In this review, we will provide a concise summary of the molecular basis and preliminary clinical outcomes of BET inhibitors in cancer therapy, with special focus on triple negative breast cancer.


PLoS Genetics ◽  
2021 ◽  
Vol 17 (10) ◽  
pp. e1009888
Author(s):  
Bin Zhu ◽  
Linhong Li ◽  
Rui Wei ◽  
Pei Liang ◽  
Xiwu Gao

The evolution of resistance to insecticides is well known to be closely associated with the overexpression of detoxifying enzymes. Although the role of glutathione S-transferase (GST) genes in insecticide resistance has been widely reported, the underlying regulatory mechanisms are poorly understood. Here, one GST gene (GSTu1) and its antisense transcript (lnc-GSTu1-AS) were identified and cloned, and both of them were upregulated in several chlorantraniliprole-resistant Plutella xylostella populations. GSTu1 was confirmed to be involved in chlorantraniliprole resistance by direct degradation of this insecticide. Furthermore, we demonstrated that lnc-GSTu1-AS interacted with GSTu1 by forming an RNA duplex, which masked the binding site of miR-8525-5p at the GSTu1-3′UTR. In summary, we revealed that lnc-GSTu1-AS maintained the mRNA stability of GSTu1 by preventing its degradation that could have been induced by miR-8525-5p and thus increased the resistance of P. xylostella to chlorantraniliprole. Our findings reveal a new noncoding RNA-mediated pathway that regulates the expression of detoxifying enzymes in insecticide-resistant insects and offer opportunities for the further understanding of the mechanisms of insecticide and drug resistance.


2019 ◽  
Vol 16 (4) ◽  
pp. 392-399 ◽  
Author(s):  
Jacob Peedicayil

Background: The rapid and major advances being made in epigenetics are impacting pharmacology, giving rise to new sub-disciplines in pharmacology, pharmacoepigenetics, the study of the epigenetic basis of variation in response to drugs; and pharmacoepigenomics, the application of pharmacoepigenetics on a genome-wide scale. Methods: This article highlights the following aspects of pharmacoepigenetics and pharmacoepigenomics: epigenetic therapy, the role of epigenetics in pharmacokinetics, the relevance of epigenetics to adverse drug reactions, personalized medicine, drug addiction, and drug resistance, and the use of epigenetic biomarkers in drug therapy. Results: Epigenetics is having an increasing impact on several areas of pharmacology. Conclusion: Pharmacoepigenetics and pharmacoepigenomics are new sub-disciplines in pharmacology and are likely to have an increasing impact on the use of drugs in clinical practice.


2020 ◽  
Vol 21 (16) ◽  
pp. 5758 ◽  
Author(s):  
Giulia Poli ◽  
Consuelo Fabi ◽  
Marina Maria Bellet ◽  
Claudio Costantini ◽  
Luisa Nunziangeli ◽  
...  

The innate immune system represents the host’s first-line defense against pathogens, dead cells or environmental factors. One of the most important inflammatory pathways is represented by the activation of the NOD-like receptor (NLR) protein family. Some NLRs induce the assembly of large caspase-1-activating complexes called inflammasomes. Different types of inflammasomes have been identified that can respond to distinct bacterial, viral or fungal infections; sterile cell damage or other stressors, such as metabolic imbalances. Epigenetic regulation has been recently suggested to provide a complementary mechanism to control inflammasome activity. This regulation can be exerted through at least three main mechanisms, including CpG DNA methylation, histones post-translational modifications and noncoding RNA expression. The repression or promotion of expression of different inflammasomes (NLRP1, NLRP2, NLRP3, NLRP4, NLRP6, NLRP7, NLRP12 and AIM2) through epigenetic mechanisms determines the development of pathologies with variable severity. For example, our team recently explored the role of microRNAs (miRNAs) targeting and modulating the components of the inflammasome as potential biomarkers in bladder cancer and during therapy. This suggests that the epigenetic control of inflammasome-related genes could represent a potential target for further investigations of molecular mechanisms regulating inflammatory pathways.


Folia Medica ◽  
2012 ◽  
Vol 54 (2) ◽  
pp. 12-16 ◽  
Author(s):  
Nikolay T. Popov ◽  
Vili K. Stoyanova ◽  
Nadezhda P. Madzhirova ◽  
Tihomir I. Vachev

ABSTRACT Epidemiological evidence suggests that etiology of schizophrenia may involve both the influence of genetic factors specific for the individual and the impact of the environment. It is quite likely that a crucial role in the disease development is played by molecular mechanisms mediating the interaction between genes and environment. Modern research have shown that epigenetic mechanisms or chemical modifications of deoxyribonucleic acids (DNA) and histone proteins remain unstable throughout life and can be changed by environmental factors. Thus the epigenetic mechanisms outline an attractive molecular hypothesis of the environment modelling role and the environmental contribution to schizophrenia progression. We give in the present study a general outline of schizophrenia as a pathological entity and discuss the role and involvement of environment versus genetic determinant (nature versus nurture) in the pathophysiolgical processes. Additionally, we focus on DNA methylation discussing the evidence for the role of that process in schizophrenia. Thirdly, we review the post-translational histone modifications and their role in schizophrenia. These investigations might surely lead further to the development of epigenetic therapy that looks promising in regard to symptom alleviation and the disease-associated cognitive deficit.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 11-12
Author(s):  
Rucha V Modak ◽  
Sara J Gosline ◽  
Alisa Damnernsawad ◽  
Tamilla Nechiporuk ◽  
Ted Laderas ◽  
...  

Background: The treatment of acute myeloid leukemia (AML) patients is a formidable challenge, due to disease heterogeneity and the ability to acquire secondary mutations in response to treatment. As a result, persistence and expansion of residual AML clones prevents complete remission. Emerging evidence suggests that extrinsic factors from the microenvironment protect AML cells from therapy, promote disease progression and drug resistance. Previous work from our lab highlights that higher levels of proinflammatory cytokines in the microenvironment, promote AML progression. This emphasizes the rationale to implement targeted therapies that block extrinsic signaling along with tumor intrinsic changes to overcome drug resistance in AML. Methods and Results: To identify factors and pathways that promote drug sensitivity and resistance in AML, we quantified the levels of 41 cytokines and growth factors from the plasma of 350 newly diagnosed AML patient samples using a multiplex luminex assay. The same cohort were analyzed for drug response to 122 small molecule inhibitors using an ex vivo functional drug sensitivity assay. These data were integrated with the gene expression and reactome pathway data to identify differentially regulated pathways and markers for drug response. Data integration revealed that protein synthesis pathways were significantly enriched in venetoclax, (Bcl-2 inhibitor) resistant AML samples, whereas, cytokine and immune pathways such as aberrant activation of monocyte chemoattractant protein-1 (MCP-1; p= 4.9e-07) were correlated with resistance to trametinib, a MEK inhibitor. In contrast, trametinib sensitivity was associated with activation of IL-1 and TLR signaling, suggesting specific extrinsic pathways may drive specific drug response. As a proof-of-concept, we dissected the role of MCP-1 activation in mediating trametinib resistance in AML. For this, we treated primary AML cells with MCP-1 in the presence of trametinib that led to a 4-fold increase in IC50 (0.4 vs 2.4 µM). To model drug response in vitro, we generated trametinib resistant AML cell lines MOLM13 and MV4;11 (FLT3-ITD mutated) and OCI-AML2 (DNMT3A mutated) by culturing cells in trametinib continuously over 4 months. All the trametinib resistant AML cell lines showed 2-10-fold increase in MCP-1 levels in whole cell lysate and in the conditioned media compared to the parental cells. Further, a long-term exposure of AML cells to MCP-1 in the presence of trametinib conferred a growth advantage with a 4-fold increase in cell numbers in comparison to the cells cultured with trametinib alone (n=4/condition). To delineate the molecular pathways driven by MCP-1 in trametinib resistance, we performed global phosphoproteomics using AML cell lines. We identified that MCP-1 activated various pro-survival pathways such as ERK, JNK, SRC, P70, and PKC kinases as well as cell cycle regulatory proteins as early as 5 mins in trametinib resistant cells. Targeting MCP-1 receptor-CCR2 by pharmacological inhibition, significantly reduced viability of trametinib resistant AML cells by 3-fold. The treatment with CCR2 inhibitor or trametinib individually reduced p-JNK levels by 3-fold in AML cells. A combined treatment with CCR2 inhibitor and trametinib, showed a 7-fold reduction in p-JNK levels. These data reinforced the role of MCP-1 in conferring trametinib resistance and that blockade of MCP-1 signaling re-sensitizes AML cells to trametinib. Conclusion: We show that MCP-1 augments trametinib resistance in AML by triggering novel signaling pathways. These effects can be reversed by blockade of MCP-1-CCR2 axis. Such combination treatment strategy incorporating extrinsic pathways together with the intrinsic ones would aid in re-sensitizing AML cells to therapy and represent an attractive new strategy. Our functional screen identifies that these extrinsic signals modulating drug response are unique to specific targeted therapy and should be considered when designing new therapies and clinical trials. We also created a large resource with 350 primary AML samples correlating microenvironment factors and gene expression with response to 122 small molecules, many of those which are FDA approved. This resource will help identify new drug response pathways in the context of microenvironment to design novel treatment regimens for preclinical and clinical testing and to improve AML outcomes. Disclosures Tyner: Syros: Research Funding; Agios: Research Funding; Incyte: Research Funding; Seattle Genetics: Research Funding; Takeda: Research Funding; Petra: Research Funding; Janssen: Research Funding; Constellation: Research Funding; Aptose: Research Funding; Genentech: Research Funding; Gilead: Research Funding; Array: Research Funding; AstraZeneca: Research Funding.


Author(s):  
Maria Jesus Vazquez ◽  
Silvia Daza-Dueñas ◽  
Manuel Tena-Sempere

Abstract Reproduction is an essential function for perpetuation of the species. As such, it is controlled by sophisticated regulatory mechanisms, which allow a perfect match between environmental conditions and internal cues, to ensure adequate pubertal maturation and achievement of reproductive capacity. Besides classical genetic regulatory events, mounting evidence has documented that different epigenetic mechanisms operate at different levels of the reproductive axis to finely tune the development and function of this complex neuroendocrine system along the lifespan. In this mini-review, we will summarize recent evidence on the role of epigenetics in the control of reproduction, with special focus on the modulation of the central components of this axis. Particular attention will be paid to the epigenetic control of puberty and Kiss1 neurons, as major developments have taken place in this domain recently. In addition, the putative role of central epigenetic mechanisms in mediating the influence of nutritional and environmental cues on reproductive function will be also discussed.


2018 ◽  
Vol 475 (14) ◽  
pp. 2305-2328 ◽  
Author(s):  
Yalda Hekmatshoar ◽  
Jean Nakhle ◽  
Mireille Galloni ◽  
Marie-Luce Vignais

Intercellular communications play a major role in tissue homeostasis. In pathologies such as cancer, cellular interactions within the tumor microenvironment (TME) contribute to tumor progression and resistance to therapy. Tunneling nanotubes (TNTs) are newly discovered long-range intercellular connections that allow the exchange between cells of various cargos, ranging from ions to whole organelles such as mitochondria. TNT-transferred mitochondria were shown to change the metabolism and functional properties of recipient cells as reported for both normal and cancer cells. Metabolic plasticity is now considered a hallmark of cancer as it notably plays a pivotal role in drug resistance. The acquisition of cancer drug resistance was also associated to TNT-mediated mitochondria transfer, a finding that relates to the role of mitochondria as a hub for many metabolic pathways. In this review, we first give a brief overview of the various mechanisms of drug resistance and of the cellular communication means at play in the TME, with a special focus on the recently discovered TNTs. We further describe recent studies highlighting the role of the TNT-transferred mitochondria in acquired cancer cell drug resistance. We also present how changes in metabolic pathways, including glycolysis, pentose phosphate and lipid metabolism, are linked to cancer cell resistance to therapy. Finally, we provide examples of novel therapeutic strategies targeting mitochondria and cell metabolism as a way to circumvent cancer cell drug resistance.


Sign in / Sign up

Export Citation Format

Share Document