1
|
Yu M, Du H, Zhang C, Shi Y. miR-192 family in breast cancer: Regulatory mechanisms and diagnostic value. Biomed Pharmacother 2024; 175:116620. [PMID: 38653113 DOI: 10.1016/j.biopha.2024.116620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
There is a growing interest in the role of the miRNA family in human cancer. The miRNA-192 family is a group of conserved small RNAs, including miR-192, miR-194, and miR-215. Recent studies have shown that the incidence and mortality of breast cancer have been increasing epidemiologically year by year, and it is urgent to clarify the pathogenesis of breast cancer and seek new diagnostic and therapeutic methods. There is increasing evidence that miR-192 family members may be involved in the occurrence and development of breast cancer. This review describes the regulatory mechanism of the miRNA-192 family affecting the malignant behavior of breast cancer cells and evaluates the value of the miRNA-192 family as a diagnostic and prognostic biomarker for breast cancer. It is expected that summarizing and discussing the relationship between miRNA-192 family members and breast cancer, it will provide a new direction for the clinical diagnosis and treatment of breast cancer and basic medical research.
Collapse
Affiliation(s)
- Mingxuan Yu
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, PR China.
| | - Hua Du
- College of Basic Medicine, Inner Mongolia Medical University, PR China; Department of Pathology, Affiliated Hospital of Inner Mongolia Medical University, PR China.
| | - Caihong Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, PR China.
| | - Yingxu Shi
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, PR China.
| |
Collapse
|
2
|
Tripathi S, Mathaiyan J, Kayal S, Nachiappa Ganesh R. The Role of Circulating MicroRNAs in the Prediction of Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer in the Indian Population. Cureus 2024; 16:e59553. [PMID: 38832155 PMCID: PMC11144582 DOI: 10.7759/cureus.59553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) are known to play an important role in cancer cell proliferation, susceptibility of cancer cells to chemotherapy, and patient survival. Identifying miRNAs that can predict response to chemotherapy in locally advanced breast cancer (LABC), the most common variant, can help to choose appropriate drug regimens to suit the epigenetic profile of individual patients. OBJECTIVE To investigate the expression of the differentially expressed miRNAs identified by next-generation sequencing from a pilot study involving cases and controls, in peripheral blood mononuclear cells (PBMC) of patients with LABC during the course of neoadjuvant chemotherapy (NAC) and determine their role in response to chemotherapy. METHODS This study included 30 newly diagnosed LABC patients. Peripheral blood from every participant was collected before the start of chemotherapy, at the end of the third cycle, and at the end of the seventh cycle of NAC. Based on the results of a pilot study in a similar population with suitable controls, four differentially expressed miRNAs namely miR-24-2, miR-192-5p, miR-3609, and miR-664b-3p were considered to be validated in this study. The expression of these four miRNAs was examined by qRT-PCR, and their association with response to chemotherapy was analyzed. RESULT A significant change in the expression of miR-192-5p was found in responders (p = 0.001) over a period of seven cycles and the difference between the expression of miR-24-2 from baseline to the seventh cycle of NAC was higher in responders while compared to the non-responders (p < 0.05). CONCLUSION miR-192-5p and miR-24-2 were identified as predictive biomarkers for response to NAC in south Indian patients with LABC.
Collapse
Affiliation(s)
- Shyam Tripathi
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Jayanthi Mathaiyan
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Smita Kayal
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Rajesh Nachiappa Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| |
Collapse
|
3
|
Waly OM, El-Mahdy NA, El-Shitany NAEA, Mohammedsaleh ZM, El-Kadem AH. Protective role of naftidrofuryl against methotrexate-induced testicular damage via the amelioration of the p53/miRNA-29a/CDC42 apoptotic pathway, inflammation, and oxidative stress. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104067. [PMID: 36649853 DOI: 10.1016/j.etap.2023.104067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/17/2022] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
This study aimed to assess the possible protective effects of naftidrofuryl (Naf) against methotrexate (MTX)-induced testicular toxicity in rats. Male rats were randomly distributed into four groups: control, Naf, MTX, and MTX+Naf groups. MTX administration induced oxidative stress, inflammation, and apoptosis in the testicular tissue, while pretreatment with Naf attenuated these pathways. Naf pretreatment significantly decreased malondialdehyde and interleukin-6 contents, microRNA-29a (miRNA-29a) expression level, and nuclear factor kappa B and p53 immunostaining in the testicular tissues compared to the MTX group. Conversely, it significantly increased Johnsen's score, serum testosterone level, serum total antioxidant capacity, testicular superoxide dismutase activity, testicular catalase activity, and testicular cell division cycle 42 (CDC42) expression compared to the MTX group. In conclusion, Naf exerted a significant protective effect against MTX-induced testicular toxicity via antioxidant and anti-inflammatory mechanisms and modulating the p53/miRNA-29a/CDC42 apoptotic pathway.
Collapse
Affiliation(s)
- Ola Mahmoud Waly
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Nageh Ahmed El-Mahdy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Aya Hassan El-Kadem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
4
|
Wang P, Wang HY, Gao XJ, Zhu HX, Zhang XP, Liu F, Wang W. Encoding and Decoding of p53 Dynamics in Cellular Response to Stresses. Cells 2023; 12:cells12030490. [PMID: 36766831 PMCID: PMC9914463 DOI: 10.3390/cells12030490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
In the cellular response to stresses, the tumor suppressor p53 is activated to maintain genomic integrity and fidelity. As a transcription factor, p53 exhibits rich dynamics to allow for discrimination of the type and intensity of stresses and to direct the selective activation of target genes involved in different processes including cell cycle arrest and apoptosis. In this review, we focused on how stresses are encoded into p53 dynamics and how the dynamics are decoded into cellular outcomes. Theoretical modeling may provide a global view of signaling in the p53 network by coupling the encoding and decoding processes. We discussed the significance of modeling in revealing the mechanisms of the transition between p53 dynamic modes. Moreover, we shed light on the crosstalk between the p53 network and other signaling networks. This review may advance the understanding of operating principles of the p53 signaling network comprehensively and provide insights into p53 dynamics-based cancer therapy.
Collapse
Affiliation(s)
- Ping Wang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Key Laboratory of High Performance Scientific Computation, School of Science, Xihua University, Chengdu 610039, China
| | - Hang-Yu Wang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Xing-Jie Gao
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Hua-Xia Zhu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- Correspondence: (X.-P.Z.); (W.W.)
| | - Feng Liu
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructure, Nanjing University, Nanjing 210093, China
- Department of Physics, Nanjing University, Nanjing 210093, China
| | - Wei Wang
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructure, Nanjing University, Nanjing 210093, China
- Department of Physics, Nanjing University, Nanjing 210093, China
- Correspondence: (X.-P.Z.); (W.W.)
| |
Collapse
|
5
|
microRNA-Mediated Encoding and Decoding of Time-Dependent Signals in Tumorigenesis. Biomolecules 2022; 12:biom12020213. [PMID: 35204714 PMCID: PMC8961662 DOI: 10.3390/biom12020213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
microRNAs, pivotal post-transcriptional regulators of gene expression, in the past decades have caught the attention of researchers for their involvement in different biological processes, ranging from cell development to cancer. Although lots of effort has been devoted to elucidate the topological features and the equilibrium properties of microRNA-mediated motifs, little is known about how the information encoded in frequency, amplitude, duration, and other features of their regulatory signals can affect the resulting gene expression patterns. Here, we review the current knowledge about microRNA-mediated gene regulatory networks characterized by time-dependent input signals, such as pulses, transient inputs, and oscillations. First, we identify the general characteristic of the main motifs underlying temporal patterns. Then, we analyze their impact on two commonly studied oncogenic networks, showing how their dysfunction can lead to tumorigenesis.
Collapse
|
6
|
Visser H, Thomas AD. MicroRNAs and the DNA damage response: How is cell fate determined? DNA Repair (Amst) 2021; 108:103245. [PMID: 34773895 DOI: 10.1016/j.dnarep.2021.103245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
It is becoming clear that the DNA damage response orchestrates an appropriate response to a given level of DNA damage, whether that is cell cycle arrest and repair, senescence or apoptosis. It is plausible that the alternative regulation of the DNA damage response (DDR) plays a role in deciding cell fate following damage. MicroRNAs (miRNAs) are associated with the transcriptional regulation of many cellular processes. They have diverse functions, affecting, presumably, all aspects of cell biology. Many have been shown to be DNA damage inducible and it is conceivable that miRNA species play a role in deciding cell fate following DNA damage by regulating the expression and activation of key DDR proteins. From a clinical perspective, miRNAs are attractive targets to improve cancer patient outcomes to DNA-damaging chemotherapy. However, cancer tissue is known to be, or to become, well adapted to DNA damage as a means of inducing chemoresistance. This frequently results from an altered DDR, possibly owing to miRNA dysregulation. Though many studies provide an overview of miRNAs that are dysregulated within cancerous tissues, a tangible, functional association is often lacking. While miRNAs are well-documented in 'ectopic biology', the physiological significance of endogenous miRNAs in the context of the DDR requires clarification. This review discusses miRNAs of biological relevance and their role in DNA damage response by potentially 'fine-tuning' the DDR towards a particular cell fate in response to DNA damage. MiRNAs are thus potential therapeutic targets/strategies to limit chemoresistance, or improve chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Hartwig Visser
- Centre for Research in Biosciences, University of the West of England, Frenchay Campus, Bristol BS16 1QY, United Kingdom
| | - Adam D Thomas
- Centre for Research in Biosciences, University of the West of England, Frenchay Campus, Bristol BS16 1QY, United Kingdom.
| |
Collapse
|
7
|
Quarton T, Kang T, Papakis V, Nguyen K, Nowak C, Li Y, Bleris L. Uncoupling gene expression noise along the central dogma using genome engineered human cell lines. Nucleic Acids Res 2020; 48:9406-9413. [PMID: 32810265 PMCID: PMC7498316 DOI: 10.1093/nar/gkaa668] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 01/22/2023] Open
Abstract
Eukaryotic protein synthesis is an inherently stochastic process. This stochasticity stems not only from variations in cell content between cells but also from thermodynamic fluctuations in a single cell. Ultimately, these inherently stochastic processes manifest as noise in gene expression, where even genetically identical cells in the same environment exhibit variation in their protein abundances. In order to elucidate the underlying sources that contribute to gene expression noise, we quantify the contribution of each step within the process of protein synthesis along the central dogma. We uncouple gene expression at the transcriptional, translational, and post-translational level using custom engineered circuits stably integrated in human cells using CRISPR. We provide a generalized framework to approximate intrinsic and extrinsic noise in a population of cells expressing an unbalanced two-reporter system. Our decomposition shows that the majority of intrinsic fluctuations stem from transcription and that coupling the two genes along the central dogma forces the fluctuations to propagate and accumulate along the same path, resulting in increased observed global correlation between the products.
Collapse
Affiliation(s)
- Tyler Quarton
- Bioengineering Department, University of Texas at Dallas, Richardson, TX, USA.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA
| | - Taek Kang
- Bioengineering Department, University of Texas at Dallas, Richardson, TX, USA.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA
| | - Vasileios Papakis
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA.,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Khai Nguyen
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA.,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Chance Nowak
- Bioengineering Department, University of Texas at Dallas, Richardson, TX, USA.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA.,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Yi Li
- Bioengineering Department, University of Texas at Dallas, Richardson, TX, USA.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA
| | - Leonidas Bleris
- Bioengineering Department, University of Texas at Dallas, Richardson, TX, USA.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA.,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
8
|
Zhang Y, Liu H, Li Z, Miao Z, Zhou J. Oscillatory Dynamics of p53-Mdm2 Circuit in Response to DNA Damage Caused by Ionizing Radiation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1703-1713. [PMID: 30762566 DOI: 10.1109/tcbb.2019.2899574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Although the dynamical behavior of the p53-Mdm2 loop has been extensively studied, the understanding of the mechanism underlying the regulation of this pathway still remains limited. Herein, we developed an integrated model with five basic components and three ubiquitous time delays for the p53-Mdm2 interaction in response to DNA damage following ionizing radiation (IR). We showed that a sufficient amount of activated ATM level can initiate the p53 oscillations with nearly the same amplitude over a wide range of the ATM level; a proper range of p53 level is also required for generating the oscillations, for too high or too low levels it would fail to generate the oscillations; and increased Mdm2 level leads to decreased amplitude of the p53 oscillation and reduced expression of the p53 activity. Moreover, we found that the negative feedback loop formed between p53 and nuclear Mdm2 plays a dominant role in determining the p53 dynamics, whereas when interaction strength of the negative feedback loop becomes weaker, the positive feedback loop formed between p53 and cytoplasmatic Mdm2 can induce different types of dynamics. Furthermore, we demonstrated that the total time delay required for protein production and nuclear translocation of Mdm2 can induce p53 oscillations even when the p53 level is at a certain stable high steady state or at a certain stable low steady state. In addition, the two important features of the oscillatory dynamics-amplitude and period-can be controlled by such time delay. These results are in agreement with multiple experimental observations and may enrich our understanding of the dynamics of the p53 network.
Collapse
|
9
|
Effects of Mycoplasmas on the Host Cell Signaling Pathways. Pathogens 2020; 9:pathogens9040308. [PMID: 32331465 PMCID: PMC7238135 DOI: 10.3390/pathogens9040308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022] Open
Abstract
Mycoplasmas are the smallest free-living organisms. Reduced sizes of their genomes put constraints on the ability of these bacteria to live autonomously and make them highly dependent on the nutrients produced by host cells. Importantly, at the organism level, mycoplasmal infections may cause pathological changes to the host, including cancer and severe immunological reactions. At the molecular level, mycoplasmas often activate the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) inflammatory response and concomitantly inhibit the p53-mediated response, which normally triggers the cell cycle and apoptosis. Thus, mycoplasmal infections may be considered as cancer-associated factors. At the same time, mycoplasmas through their membrane lipoproteins (LAMPs) along with lipoprotein derivatives (lipopeptide MALP-2, macrophage-activating lipopeptide-2) are able to modulate anti-inflammatory responses via nuclear translocation and activation of Nrf2 (the nuclear factor-E2-related anti-inflammatory transcription factor 2). Thus, interactions between mycoplasmas and host cells are multifaceted and depend on the cellular context. In this review, we summarize the current information on the role of mycoplasmas in affecting the host’s intracellular signaling mediated by the interactions between transcriptional factors p53, Nrf2, and NF-κB. A better understanding of the mechanisms underlying pathologic processes associated with reprogramming eukaryotic cells that arise during the mycoplasma-host cell interaction should facilitate the development of new therapeutic approaches to treat oncogenic and inflammatory processes.
Collapse
|
10
|
Dynamic Behavior of p53 Driven by Delay and a Microrna-34a-Mediated Feedback Loop. Int J Mol Sci 2020; 21:ijms21041271. [PMID: 32070036 PMCID: PMC7072921 DOI: 10.3390/ijms21041271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/30/2020] [Accepted: 02/09/2020] [Indexed: 01/02/2023] Open
Abstract
The tumor suppressor protein p53 is a critical hub in the comprehensive transcriptional network that inhibits the growth of cells after acute stress stimulation. In this paper, an integrated model of the p53 signaling pathway in response to DNA damage is proposed and the p53 stability and oscillatory dynamics are analyzed. Through theoretical analysis and numerical simulation, we find that the delay as a bifurcation parameter can drive the p53-Mdm2 module to undergo a supercritical Hopf bifurcation, thereby producing oscillation behavior. Moreover, we demonstrate how the positive feedback loop formed by p53* and microRNA-34a (miR-34a) with the feature of double-negative regulation produces limit-cycle oscillations. Further, we find that miR-34a can affect the critical value of Hopf bifurcation in delay-induced p53 networks. In addition, we show that ATM, once activated by DNA damage, makes p53* undergo two Hopf bifurcations. These results revealed that both time delay and miR-34a can have tumor suppressing roles by promoting p53 oscillation or high level expression, which will provide a perspective for promoting the development of anti-cancer drugs by targeting miR-34a and time delay.
Collapse
|
11
|
Ferro E, Enrico Bena C, Grigolon S, Bosia C. From Endogenous to Synthetic microRNA-Mediated Regulatory Circuits: An Overview. Cells 2019; 8:E1540. [PMID: 31795372 PMCID: PMC6952906 DOI: 10.3390/cells8121540] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs are short non-coding RNAs that are evolutionarily conserved and are pivotal post-transcriptional mediators of gene regulation. Together with transcription factors and epigenetic regulators, they form a highly interconnected network whose building blocks can be classified depending on the number of molecular species involved and the type of interactions amongst them. Depending on their topology, these molecular circuits may carry out specific functions that years of studies have related to the processing of gene expression noise. In this review, we first present the different over-represented network motifs involving microRNAs and their specific role in implementing relevant biological functions, reviewing both theoretical and experimental studies. We then illustrate the recent advances in synthetic biology, such as the construction of artificially synthesised circuits, which provide a controlled tool to test experimentally the possible microRNA regulatory tasks and constitute a starting point for clinical applications.
Collapse
Affiliation(s)
- Elsi Ferro
- IIGM—Italian Institute for Genomic Medicine, c/o IRCCS, 10060 Candiolo (Torino), Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo (Torino), Italy
| | - Chiara Enrico Bena
- IIGM—Italian Institute for Genomic Medicine, c/o IRCCS, 10060 Candiolo (Torino), Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo (Torino), Italy
| | - Silvia Grigolon
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Carla Bosia
- IIGM—Italian Institute for Genomic Medicine, c/o IRCCS, 10060 Candiolo (Torino), Italy
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
12
|
Kim E, Kim JY, Lee JY. Mathematical Modeling of p53 Pathways. Int J Mol Sci 2019; 20:ijms20205179. [PMID: 31635420 PMCID: PMC6834204 DOI: 10.3390/ijms20205179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cells have evolved balanced systems that ensure an appropriate response to stress. The systems elicit repair responses in temporary or moderate stress but eliminate irreparable cells via apoptosis in detrimental conditions of prolonged or severe stress. The tumor suppressor p53 is a central player in these stress response systems. When activated under DNA damage stress, p53 regulates hundreds of genes that are involved in DNA repair, cell cycle, and apoptosis. Recently, increasing studies have demonstrated additional regulatory roles of p53 in metabolism and mitochondrial physiology. Due to the inherent complexity of feedback loops between p53 and its target genes, the application of mathematical modeling has emerged as a novel approach to better understand the multifaceted functions and dynamics of p53. In this review, we discuss several mathematical modeling approaches in exploring the p53 pathways.
Collapse
Affiliation(s)
- Eunjung Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| | - Joo-Yong Lee
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Korea.
- Korea Basic Science Institute, Daejeon 34133, Korea.
| |
Collapse
|
13
|
Sun CY, Zhang XP, Wang W. Coordination of miR-192 and miR-22 in p53-Mediated Cell Fate Decision. Int J Mol Sci 2019; 20:ijms20194768. [PMID: 31561425 PMCID: PMC6801623 DOI: 10.3390/ijms20194768] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/08/2019] [Accepted: 09/22/2019] [Indexed: 12/14/2022] Open
Abstract
p53-targeted microRNAs (miRNAs) markedly affect cellular response to DNA damage. These miRNAs may contribute to either cell cycle arrest or apoptosis induction. However, how these miRNAs coordinate to modulate the decision between cell survival and death remains less understood. Here, we developed an integrated model of p53 signaling network to investigate how p53-targeted miR-192 and miR-22 modulate cellular outcome in response to DNA damage. By numerical simulations, we found that p53 is activated progressively depending on the extent of DNA damage. Upon moderate damage, p53 rises to medium levels and induces miR-192 to promote its own activation, facilitating p21 induction and cell cycle arrest. Upon severe damage, p53 reaches high levels and is fully activated due to phosphatase and tensin homolog (PTEN) induction. As a result, it transactivates miR-22 to repress p21 expression and activate E2F1, resulting in apoptosis. Therefore, miR-192 promotes primary activation of p53, while miR-22 promotes apoptosis by downregulating p21. This work may advance the understanding of the mechanism for cell fate decision between life and death by p53-inducible miRNAs.
Collapse
Affiliation(s)
- Cheng-Yuan Sun
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, Nanjing 210093, China.
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China.
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China.
| | - Wei Wang
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, Nanjing 210093, China.
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
14
|
Li Z, Liu S, Yang Q. Incoherent Inputs Enhance the Robustness of Biological Oscillators. Cell Syst 2019; 5:72-81.e4. [PMID: 28750200 DOI: 10.1016/j.cels.2017.06.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/30/2017] [Accepted: 06/22/2017] [Indexed: 11/25/2022]
Abstract
Robust biological oscillators retain the critical ability to function in the presence of environmental perturbations. Although central architectures that support robust oscillations have been extensively studied, networks containing the same core vary drastically in their potential to oscillate, and it remains elusive what peripheral modifications to the core contribute to this functional variation. Here, we have generated a complete atlas of two- and three-node oscillators computationally, then systematically analyzed the association between network structure and robustness. We found that, while certain core topologies are essential for producing a robust oscillator, local structures can substantially modulate the robustness of oscillations. Notably, local nodes receiving incoherent or coherent inputs respectively promote or attenuate the overall network robustness in an additive manner. We validated these relationships in larger-scale networks reflective of real biological oscillators. Our findings provide an explanation for why auxiliary structures not required for oscillation are evolutionarily conserved and suggest simple ways to evolve or design robust oscillators.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Shixuan Liu
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Qiong Yang
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Guha A, Ahuja D, Das Mandal S, Parasar B, Deyasi K, Roy D, Sharma V, Willard B, Ghosh A, Ray PS. Integrated Regulation of HuR by Translation Repression and Protein Degradation Determines Pulsatile Expression of p53 Under DNA Damage. iScience 2019; 15:342-359. [PMID: 31103853 PMCID: PMC6548907 DOI: 10.1016/j.isci.2019.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/20/2018] [Accepted: 05/01/2019] [Indexed: 12/21/2022] Open
Abstract
Expression of tumor suppressor p53 is regulated at multiple levels, disruption of which often leads to cancer. We have adopted an approach combining computational systems modeling with experimental validation to elucidate the translation regulatory network that controls p53 expression post DNA damage. The RNA-binding protein HuR activates p53 mRNA translation in response to UVC-induced DNA damage in breast carcinoma cells. p53 and HuR levels show pulsatile change post UV irradiation. The computed model fitted with the observed pulse of p53 and HuR only when hypothetical regulators of synthesis and degradation of HuR were incorporated. miR-125b, a UV-responsive microRNA, was found to represses the translation of HuR mRNA. Furthermore, UV irradiation triggered proteasomal degradation of HuR mediated by an E3-ubiquitin ligase tripartite motif-containing 21 (TRIM21). The integrated action of miR-125b and TRIM21 constitutes an intricate control system that regulates pulsatile expression of HuR and p53 and determines cell viability in response to DNA damage.
Collapse
Affiliation(s)
- Abhishek Guha
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Deepika Ahuja
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Sukhen Das Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Bibudha Parasar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Krishanu Deyasi
- Department of Mathematics and Statistics, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Debadrita Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Vasundhara Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Anandamohan Ghosh
- Department of Physical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Partho Sarothi Ray
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India.
| |
Collapse
|
16
|
Central role of the p53 pathway in the noncoding-RNA response to oxidative stress. Aging (Albany NY) 2019; 9:2559-2586. [PMID: 29242407 PMCID: PMC5764393 DOI: 10.18632/aging.101341] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022]
Abstract
Oxidative stress plays a fundamental role in many conditions. Specifically, redox imbalance inhibits endothelial cell (EC) growth, inducing cell death and senescence. We used global transcriptome profiling to investigate the involvement of noncoding-RNAs in these phenotypes. By RNA-sequencing, transcriptome changes were analyzed in human ECs exposed to H2O2, highlighting a pivotal role of p53-signaling. Bioinformatic analysis and validation in p53-silenced ECs, identified several p53-targets among both mRNAs and long noncoding-RNAs (lncRNAs), including MALAT1 and NEAT1. Among microRNAs (miRNAs), miR-192-5p was the most induced by H2O2 treatment, in a p53-dependent manner. Down-modulated mRNA-targets of miR-192-5p were involved in cell cycle, DNA repair and stress response. Accordingly, miR-192-5p overexpression significantly decreased EC proliferation, inducing cell death. A central role of the p53-pathway was also confirmed by the analysis of differential exon usage: Upon H2O2 treatment, the expression of p53-dependent 5'-isoforms of MDM2 and PVT1 increased selectively. The transcriptomic alterations identified in H2O2-treated ECs were also observed in other physiological and pathological conditions where redox control plays a fundamental role, such as ECs undergoing replicative senescence, skeletal muscles of critical limb-ischemia patients and the peripheral-blood mononuclear cells of long-living individuals. Collectively, these findings indicate a prominent role of noncoding-RNAs in oxidative stress response.
Collapse
|
17
|
Zhao C, Zhang Y, Popel AS. Mechanistic Computational Models of MicroRNA-Mediated Signaling Networks in Human Diseases. Int J Mol Sci 2019; 20:E421. [PMID: 30669429 PMCID: PMC6358731 DOI: 10.3390/ijms20020421] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRs) are endogenous non-coding RNA molecules that play important roles in human health and disease by regulating gene expression and cellular processes. In recent years, with the increasing scientific knowledge and new discovery of miRs and their gene targets, as well as the plentiful experimental evidence that shows dysregulation of miRs in a wide variety of human diseases, the computational modeling approach has emerged as an effective tool to help researchers identify novel functional associations between differential miR expression and diseases, dissect the phenotypic expression patterns of miRs in gene regulatory networks, and elucidate the critical roles of miRs in the modulation of disease pathways from mechanistic and quantitative perspectives. Here we will review the recent systems biology studies that employed different kinetic modeling techniques to provide mechanistic insights relating to the regulatory function and therapeutic potential of miRs in human diseases. Some of the key computational aspects to be discussed in detail in this review include (i) models of miR-mediated network motifs in the regulation of gene expression, (ii) models of miR biogenesis and miR⁻target interactions, and (iii) the incorporation of such models into complex disease pathways in order to generate mechanistic, molecular- and systems-level understanding of pathophysiology. Other related bioinformatics tools such as computational platforms that predict miR-disease associations will also be discussed, and we will provide perspectives on the challenges and opportunities in the future development and translational application of data-driven systems biology models that involve miRs and their regulatory pathways in human diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
XPD suppresses cell proliferation and migration via miR-29a-3p-Mdm2/PDGF-B axis in HCC. Cell Biosci 2019; 9:6. [PMID: 30627419 PMCID: PMC6321695 DOI: 10.1186/s13578-018-0269-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/31/2018] [Indexed: 02/08/2023] Open
Abstract
Objective The aim of this study was to investigate the role of XPD in migration and invasion of hepatocellular carcinoma (HCC) cells. Methods The expression of XPD and miR-29a-3p was examined by western blot and qRT-PCR, cell proliferation was detected by MTT assay, cell migration was detected by transwell assay. TargetScan was used to predict potential targets of miR-29a-3p. Results In this study, we found that the expression of XPD and miR-29a-3p was downregulated in HCC samples and HCC cell lines. XPD suppressed proliferation and migration of HCC cell via regulating miR-29a-3p expression. Target prediction analysis and dual-luciferase reporter assay confirmed Mdm2 and PDGF-B were direct targets of miR-29a-3p, and miR-29a-3p suppressed proliferation and migration of HCC cells via regulating the expression of Mdm2 or PDGF-B. Conclusions Our data indicated that XPD suppressed cell proliferation and migration via miR-29a-3p-Mdm2/PDGF-B axis in HCC.
Collapse
|
19
|
Chen ZJ, Yan YJ, Shen H, Zhou JJ, Yang GH, Liao YX, Zeng JM, Yang T. miR-192 Is Overexpressed and Promotes Cell Proliferation in Prostate Cancer. Med Princ Pract 2019; 28:124-132. [PMID: 30544100 PMCID: PMC6546031 DOI: 10.1159/000496206] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Prostate cancer (PCa) is one of the most prevalent types of cancer among men worldwide. The incidence of PCa is increasing in China. Therefore, there is an urgent need to identify novel diagnostic and prognostic markers for PCa to improve the treatment of the disease. METHODS The Cancer Genome Atlas (TCGA) and GEO database were used to analyze the expression of miR-192, and the relationship between miR-192 and the clinical features of patients with PCa. Cell cycle and cell proliferation assay were used to detect the functional roles of miR-192 in PCa. Bioinformatic analysis for miR-192-5p was performed using gene ontology and KEGG analysis. RESULTS By analyzing the dataset of TCGA, we found that miR-192 was overexpressed in PCa samples compared to normal tissues and was upregulated in high-grade PCa compared to low-grade PCa. We also observed that higher miR-192 expression was associated with a shorter biochemical recurrence-free survival time. Our results also demonstrated that miR-192 promoted PCa cell proliferation and cell cycle progression. CONCLUSION These results suggest that miR-192 may be considered for use as a potential diagnostic and therapeutic target of PCa.
Collapse
Affiliation(s)
- Zhong-Jun Chen
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - You-Ji Yan
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Hao Shen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China,
| | - Jia-Jie Zhou
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Guang-Hua Yang
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Yi-Xiang Liao
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Jin-Min Zeng
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Tao Yang
- Department of Urology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| |
Collapse
|
20
|
Borchsenius SN, Daks A, Fedorova O, Chernova O, Barlev NA. Effects of mycoplasma infection on the host organism response via p53/NF‐κB signaling. J Cell Physiol 2018; 234:171-180. [DOI: 10.1002/jcp.26781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022]
Affiliation(s)
| | - Alexandra Daks
- Institute of Cytology RAS, Laboratory of Gene Expression Regulation Saint‐Petersburg Russia
| | - Olga Fedorova
- Institute of Cytology RAS, Laboratory of Gene Expression Regulation Saint‐Petersburg Russia
| | - Olga Chernova
- Kazan Scientific Center Kazan Institute of Biochemistry and Biophysics, Laboratory “Omics Technology”, Russian Academy of Sciences Kazan Russia
| | - Nickolai A. Barlev
- Institute of Cytology RAS, Laboratory of Gene Expression Regulation Saint‐Petersburg Russia
| |
Collapse
|
21
|
Li Y, Nowak CM, Withers D, Pertsemlidis A, Bleris L. CRISPR-Based Editing Reveals Edge-Specific Effects in Biological Networks. CRISPR J 2018; 1:286-293. [PMID: 31021219 DOI: 10.1089/crispr.2018.0018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Unraveling the properties of biological networks is central to understanding both normal and disease cellular phenotypes. Networks consist of functional elements (nodes) that form a variety of diverse connections (edges), with each node being a hub for multiple edges. Herein, in contrast to node-centric network perturbation and analysis approaches, we present a high-throughput CRISPR-based methodology for delineating the role of network edges. Ablation of network edges using a library targeting 93 miRNA target sites in 71 genes reveals numerous edges that control, with variable importance, cellular growth and survival under stress. To compare the impact of removing nodes versus edges in a biological network, we dissect a specific p53-microRNA pathway. We show that removal of the miR-34a target site from the anti-apoptotic gene BCL2 desensitizes the cell to ectopic delivery of miR-34a in a p53-dependent manner. In summary, we demonstrate that network edges are critical to the function and stability of biological networks. Our results introduce a novel genetic screening opportunity via edge ablation and highlight a new dimension in biological network analysis.
Collapse
Affiliation(s)
- Yi Li
- 1 Department of Bioengineering, The University of Texas at Dallas, Richardson, Texas.,2 Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas
| | - Chance M Nowak
- 2 Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas.,3 Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Daniel Withers
- 1 Department of Bioengineering, The University of Texas at Dallas, Richardson, Texas.,2 Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas
| | - Alexander Pertsemlidis
- 4 Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Leonidas Bleris
- 1 Department of Bioengineering, The University of Texas at Dallas, Richardson, Texas.,2 Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas.,3 Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
22
|
Quarton T, Ehrhardt K, Lee J, Kannan S, Li Y, Ma L, Bleris L. Mapping the operational landscape of microRNAs in synthetic gene circuits. NPJ Syst Biol Appl 2018; 4:6. [PMID: 29354284 PMCID: PMC5765153 DOI: 10.1038/s41540-017-0043-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/27/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are a class of short, noncoding RNAs that are ubiquitous modulators of gene expression, with roles in development, homeostasis, and disease. Engineered microRNAs are now frequently used as regulatory modules in synthetic biology. Moreover, synthetic gene circuits equipped with engineered microRNA targets with perfect complementarity to endogenous microRNAs establish an interface with the endogenous milieu at the single-cell level. The function of engineered microRNAs and sensor systems is typically optimized through extensive trial-and-error. Here, using a combination of synthetic biology experimentation in human embryonic kidney cells and quantitative analysis, we investigate the relationship between input genetic template abundance, microRNA concentration, and output under microRNA control. We provide a framework that employs the complete operational landscape of a synthetic gene circuit and enables the stepwise development of mathematical models. We derive a phenomenological model that recapitulates experimentally observed nonlinearities and contains features that provide insight into the microRNA function at various abundances. Our work facilitates the characterization and engineering of multi-component genetic circuits and specifically points to new insights on the operation of microRNAs as mediators of endogenous information and regulators of gene expression in synthetic biology.
Collapse
Affiliation(s)
- Tyler Quarton
- 1Bioengineering Department, University of Texas at Dallas, Richardson, TX USA.,2Center for Systems Biology, University of Texas at Dallas, Richardson, TX USA
| | - Kristina Ehrhardt
- 1Bioengineering Department, University of Texas at Dallas, Richardson, TX USA.,2Center for Systems Biology, University of Texas at Dallas, Richardson, TX USA
| | - James Lee
- 3Department of Biological Sciences, University of Texas at Dallas, Richardson, TX USA
| | - Srijaa Kannan
- 4School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX USA
| | - Yi Li
- 1Bioengineering Department, University of Texas at Dallas, Richardson, TX USA.,2Center for Systems Biology, University of Texas at Dallas, Richardson, TX USA
| | - Lan Ma
- 1Bioengineering Department, University of Texas at Dallas, Richardson, TX USA
| | - Leonidas Bleris
- 1Bioengineering Department, University of Texas at Dallas, Richardson, TX USA.,2Center for Systems Biology, University of Texas at Dallas, Richardson, TX USA.,3Department of Biological Sciences, University of Texas at Dallas, Richardson, TX USA
| |
Collapse
|
23
|
Ballweg R, Paek AL, Zhang T. A dynamical framework for complex fractional killing. Sci Rep 2017; 7:8002. [PMID: 28808338 PMCID: PMC5556027 DOI: 10.1038/s41598-017-07422-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/26/2017] [Indexed: 11/09/2022] Open
Abstract
When chemotherapy drugs are applied to tumor cells with the same or similar genotypes, some cells are killed, while others survive. This fractional killing contributes to drug resistance in cancer. Through an incoherent feedforward loop, chemotherapy drugs not only activate p53 to induce cell death, but also promote the expression of apoptosis inhibitors which inhibit cell death. Consequently, cells in which p53 is activated early undergo apoptosis while cells in which p53 is activated late survive. The incoherent feedforward loop and the essential role of p53 activation timing makes fractional killing a complex dynamical challenge, which is hard to understand with intuition alone. To better understand this process, we have constructed a representative model by integrating the control of apoptosis with the relevant signaling pathways. After the model was trained to recapture the observed properties of fractional killing, it was analyzed with nonlinear dynamical tools. The analysis suggested a simple dynamical framework for fractional killing, which predicts that cell fate can be altered in three possible ways: alteration of bifurcation geometry, alteration of cell trajectories, or both. These predicted categories can explain existing strategies known to combat fractional killing and facilitate the design of novel strategies.
Collapse
Affiliation(s)
- Richard Ballweg
- Department of Molecular and Cellular Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Andrew L Paek
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Tongli Zhang
- Department of Molecular and Cellular Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
24
|
Phillips NE, Manning C, Papalopulu N, Rattray M. Identifying stochastic oscillations in single-cell live imaging time series using Gaussian processes. PLoS Comput Biol 2017; 13:e1005479. [PMID: 28493880 PMCID: PMC5444866 DOI: 10.1371/journal.pcbi.1005479] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 05/25/2017] [Accepted: 03/24/2017] [Indexed: 12/05/2022] Open
Abstract
Multiple biological processes are driven by oscillatory gene expression at different time scales. Pulsatile dynamics are thought to be widespread, and single-cell live imaging of gene expression has lead to a surge of dynamic, possibly oscillatory, data for different gene networks. However, the regulation of gene expression at the level of an individual cell involves reactions between finite numbers of molecules, and this can result in inherent randomness in expression dynamics, which blurs the boundaries between aperiodic fluctuations and noisy oscillators. This underlies a new challenge to the experimentalist because neither intuition nor pre-existing methods work well for identifying oscillatory activity in noisy biological time series. Thus, there is an acute need for an objective statistical method for classifying whether an experimentally derived noisy time series is periodic. Here, we present a new data analysis method that combines mechanistic stochastic modelling with the powerful methods of non-parametric regression with Gaussian processes. Our method can distinguish oscillatory gene expression from random fluctuations of non-oscillatory expression in single-cell time series, despite peak-to-peak variability in period and amplitude of single-cell oscillations. We show that our method outperforms the Lomb-Scargle periodogram in successfully classifying cells as oscillatory or non-oscillatory in data simulated from a simple genetic oscillator model and in experimental data. Analysis of bioluminescent live-cell imaging shows a significantly greater number of oscillatory cells when luciferase is driven by a Hes1 promoter (10/19), which has previously been reported to oscillate, than the constitutive MoMuLV 5’ LTR (MMLV) promoter (0/25). The method can be applied to data from any gene network to both quantify the proportion of oscillating cells within a population and to measure the period and quality of oscillations. It is publicly available as a MATLAB package. Technological advances now allow us to observe gene expression in real-time at a single-cell level. In a wide variety of biological contexts this new data has revealed that gene expression is highly dynamic and possibly oscillatory. It is thought that periodic gene expression may be useful for keeping track of time and space, as well as transmitting information about signalling cues. Classifying a time series as periodic from single cell data is difficult because it is necessary to distinguish whether peaks and troughs are generated from an underlying oscillator or whether they are aperiodic fluctuations. To this end, we present a novel tool to classify live-cell data as oscillatory or non-oscillatory that accounts for inherent biological noise. We first demonstrate that the method outperforms a competing scheme in classifying computationally simulated single-cell data, and we subsequently analyse live-cell imaging time series. Our method is able to successfully detect oscillations in a known genetic oscillator, but it classifies data from a constitutively expressed gene as aperiodic. The method forms a basis for discovering new gene expression oscillators and quantifying how oscillatory activity alters in response to changes in cell fate and environmental or genetic perturbations.
Collapse
Affiliation(s)
- Nick E. Phillips
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Cerys Manning
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nancy Papalopulu
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- * E-mail: (NP); (MR)
| | - Magnus Rattray
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- * E-mail: (NP); (MR)
| |
Collapse
|
25
|
Batchelor E, Loewer A. Recent progress and open challenges in modeling p53 dynamics in single cells. ACTA ACUST UNITED AC 2017; 3:54-59. [PMID: 29062976 DOI: 10.1016/j.coisb.2017.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In mammalian cells, the tumor suppressor p53 is activated upon a variety of cellular stresses and ensures an appropriate response ranging from arrest and repair to the induction of senescence and apoptosis. Quantitative measurements in individual living cells showed stimulus-dependent dynamics of p53 accumulation upon stress induction. Due to the complexity of the underlying biochemical interactions, mathematical models were indispensable for understanding the topology of the network regulating p53 dynamics. Recent work provides furhter insights into the causes of heterogeneous responses in individual cells, the rewiring of the network in response to different inputs and the role of the downstream processes in determining the cellular fate upon stress.
Collapse
Affiliation(s)
- Eric Batchelor
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, MSC 1500, Bethesda, MD 20892, USA
| | - Alexander Loewer
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 13, 64287 Darmstadt, Germany
| |
Collapse
|
26
|
Van Roosbroeck K, Fanini F, Setoyama T, Ivan C, Rodriguez-Aguayo C, Fuentes-Mattei E, Xiao L, Vannini I, Redis RS, D'Abundo L, Zhang X, Nicoloso MS, Rossi S, Gonzalez-Villasana V, Rupaimoole R, Ferracin M, Morabito F, Neri A, Ruvolo PP, Ruvolo VR, Pecot CV, Amadori D, Abruzzo L, Calin S, Wang X, You MJ, Ferrajoli A, Orlowski R, Plunkett W, Lichtenberg TM, Davuluri RV, Berindan-Neagoe I, Negrini M, Wistuba II, Kantarjian HM, Sood AK, Lopez-Berestein G, Keating MJ, Fabbri M, Calin GA. Combining Anti-Mir-155 with Chemotherapy for the Treatment of Lung Cancers. Clin Cancer Res 2016; 23:2891-2904. [PMID: 27903673 DOI: 10.1158/1078-0432.ccr-16-1025] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/19/2016] [Accepted: 11/08/2016] [Indexed: 12/31/2022]
Abstract
Purpose: The oncogenic miR-155 is upregulated in many human cancers, and its expression is increased in more aggressive and therapy-resistant tumors, but the molecular mechanisms underlying miR-155-induced therapy resistance are not fully understood. The main objectives of this study were to determine the role of miR-155 in resistance to chemotherapy and to evaluate anti-miR-155 treatment to chemosensitize tumors.Experimental Design: We performed in vitro studies on cell lines to investigate the role of miR-155 in therapy resistance. To assess the effects of miR-155 inhibition on chemoresistance, we used an in vivo orthotopic lung cancer model of athymic nude mice, which we treated with anti-miR-155 alone or in combination with chemotherapy. To analyze the association of miR-155 expression and the combination of miR-155 and TP53 expression with cancer survival, we studied 956 patients with lung cancer, chronic lymphocytic leukemia, and acute lymphoblastic leukemia.Results: We demonstrate that miR-155 induces resistance to multiple chemotherapeutic agents in vitro, and that downregulation of miR-155 successfully resensitizes tumors to chemotherapy in vivo We show that anti-miR-155-DOPC can be considered non-toxic in vivo We further demonstrate that miR-155 and TP53 are linked in a negative feedback mechanism and that a combination of high expression of miR-155 and low expression of TP53 is significantly associated with shorter survival in lung cancer.Conclusions: Our findings support the existence of an miR-155/TP53 feedback loop, which is involved in resistance to chemotherapy and which can be specifically targeted to overcome drug resistance, an important cause of cancer-related death. Clin Cancer Res; 23(11); 2891-904. ©2016 AACR.
Collapse
Affiliation(s)
- Katrien Van Roosbroeck
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Francesca Fanini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) S.r.l. IRCCS, Unit of Gene Therapy, Meldola (FC) 47014, Italy
| | - Tetsuro Setoyama
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Enrique Fuentes-Mattei
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ivan Vannini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) S.r.l. IRCCS, Unit of Gene Therapy, Meldola (FC) 47014, Italy
| | - Roxana S Redis
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lucilla D'Abundo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Xinna Zhang
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Milena S Nicoloso
- Division of Experimental Oncology 2, CRO, National Cancer Institute, Aviano 33081, Italy
| | - Simona Rossi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vianey Gonzalez-Villasana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Departamento de Biologia Celular y Genetica, Universidad Autonoma de Nuevo Leon, 66450 San Nicolas de los Garza, Nuevo Leon, Mexico
| | - Rajesha Rupaimoole
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, Bologna 40126, Italy
| | | | - Antonino Neri
- Department of Clinical Sciences and Community Health, University of Milano and Hematology, Ospedale Policlinico, Milano 20122, Italy
| | - Peter P Ruvolo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vivian R Ruvolo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chad V Pecot
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dino Amadori
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) S.r.l. IRCCS, Unit of Gene Therapy, Meldola (FC) 47014, Italy
| | - Lynne Abruzzo
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA
| | - Steliana Calin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tara M Lichtenberg
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ramana V Davuluri
- Department of Preventive Medicine - Division of Health and Biomedical Informatics, Northwestern University - Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ioana Berindan-Neagoe
- Department of Functional Genomics, The Oncology Institute, 400015 Cluj-Napoca, Romania.,Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy Iuliu Hatieganu, 400012 Cluj-Napoca, Romania
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Ignacio I Wistuba
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K Sood
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Muller Fabbri
- Departments of Pediatrics and Molecular Microbiology & Immunology, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Saban
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
27
|
Lai X, Wolkenhauer O, Vera J. Understanding microRNA-mediated gene regulatory networks through mathematical modelling. Nucleic Acids Res 2016; 44:6019-35. [PMID: 27317695 PMCID: PMC5291278 DOI: 10.1093/nar/gkw550] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
The discovery of microRNAs (miRNAs) has added a new player to the regulation of gene expression. With the increasing number of molecular species involved in gene regulatory networks, it is hard to obtain an intuitive understanding of network dynamics. Mathematical modelling can help dissecting the role of miRNAs in gene regulatory networks, and we shall here review the most recent developments that utilise different mathematical modelling approaches to provide quantitative insights into the function of miRNAs in the regulation of gene expression. Key miRNA regulation features that have been elucidated via modelling include: (i) the role of miRNA-mediated feedback and feedforward loops in fine-tuning of gene expression; (ii) the miRNA–target interaction properties determining the effectiveness of miRNA-mediated gene repression; and (iii) the competition for shared miRNAs leading to the cross-regulation of genes. However, there is still lack of mechanistic understanding of many other properties of miRNA regulation like unconventional miRNA–target interactions, miRNA regulation at different sub-cellular locations and functional miRNA variant, which will need future modelling efforts to deal with. This review provides an overview of recent developments and challenges in this field.
Collapse
Affiliation(s)
- Xin Lai
- Laboratory of Systems Tumour Immunology, Department of Dermatology, Erlangen University Hospital and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, Rostock, 18051, Germany Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, 7600, South Africa
| | - Julio Vera
- Laboratory of Systems Tumour Immunology, Department of Dermatology, Erlangen University Hospital and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, 91054, Germany
| |
Collapse
|