Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Sci Rep ; 6: 32823, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27610602

ABSTRACT

The restriction point marks a switch in G1 from growth factor-dependent to growth factor-independent progression of the cell cycle. The proper regulation of this switch is important for normal cell processes; aberrations could result in a number of diseases such as cancer, neurodegenerative disorders, stroke and myocardial infarction. To further understand the regulation of the restriction point, we extended a mathematical model of the Rb-E2F pathway to include members of the microRNA cluster miR-17-92. Our mathematical analysis shows that microRNAs play an essential role in fine-tuning and providing robustness to the switch. We also demonstrate how microRNA regulation can steer cells in or out of cancer states.


Subject(s)
E2F Transcription Factors/genetics , MicroRNAs/genetics , Neoplasms/genetics , Retinoblastoma Protein/genetics , Cell Cycle , Gene Regulatory Networks , Humans , Models, Theoretical , Signal Transduction
2.
Math Biosci Eng ; 12(6): 1277-88, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26775863

ABSTRACT

We propose the hypothesis that for a particular type of cancer there exists a key pair of oncogene (OCG) and tumor suppressor gene (TSG) that is normally involved in strong stabilizing negative feedback loops (nFBLs) of molecular interactions, and it is these interactions that are sufficiently perturbed during cancer development. These nFBLs are thought to regulate oncogenic positive feedback loops (pFBLs) that are often required for the normal cellular functions of oncogenes. Examples given in this paper are the pairs of MYC and p53, KRAS and INK4A, and E2F1 and miR-17-92. We propose dynamical models of the aforementioned OCG-TSG interactions and derive stability conditions of the steady states in terms of strengths of cycles in the qualitative interaction network. Although these conditions are restricted to predictions of local stability, their simple linear expressions in terms of competing nFBLs and pFBLs make them intuitive and practical guides for experimentalists aiming to discover drug targets and stabilize cancer networks.


Subject(s)
Genes, Tumor Suppressor , Models, Genetic , Neoplasms/genetics , Oncogenes , Cell Differentiation/genetics , Cell Proliferation/genetics , Feedback, Physiological , Gene Regulatory Networks , Humans , Mathematical Concepts , Neoplasms/etiology , Neoplasms/pathology
3.
PLoS One ; 9(12): e116050, 2014.
Article in English | MEDLINE | ID: mdl-25545504

ABSTRACT

Accumulating data indicate that cancer stem cells contribute to tumor chemoresistance and their persistence alters clinical outcome. Our previous study has shown that ovarian cancer may be initiated by ovarian cancer initiating cells (OCIC) characterized by surface antigen CD44 and c-KIT (CD117). It has been experimentally demonstrated that a microRNA, namely miR-193a, targets c-KIT mRNA for degradation and could play a crucial role in ovarian cancer development. How miR-193a is regulated is poorly understood and the emerging picture is complex. To unravel this complexity, we propose a mathematical model to explore how estrogen-mediated up-regulation of another target of miR-193a, namely E2F6, can attenuate the function of miR-193a in two ways, one through a competition of E2F6 and c-KIT transcripts for miR-193a, and second by binding of E2F6 protein, in association with a polycomb complex, to the promoter of miR-193a to down-regulate its transcription. Our model predicts that this bimodal control increases the expression of c-KIT and that the second mode of epigenetic regulation is required to generate a switching behavior in c-KIT and E2F6 expressions. Additional analysis of the TCGA ovarian cancer dataset demonstrates that ovarian cancer patients with low expression of EZH2, a polycomb-group family protein, show positive correlation between E2F6 and c-KIT. We conjecture that a simultaneous EZH2 inhibition and anti-estrogen therapy can constitute an effective combined therapeutic strategy against ovarian cancer.


Subject(s)
Epigenesis, Genetic , MicroRNAs/genetics , Models, Genetic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Base Sequence , Cell Line, Tumor , Databases, Genetic , E2F6 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , MicroRNAs/metabolism , Molecular Sequence Data , Proto-Oncogene Proteins c-kit/metabolism , Reproducibility of Results
4.
Adv Exp Med Biol ; 774: 149-67, 2013.
Article in English | MEDLINE | ID: mdl-23377973

ABSTRACT

An increasing number of transcription factors (TFs) and microRNAs (miRNAs) is known to form feedback loops (FBLs) of interactions where a TF positively or negatively regulates the expression of a miRNA, and the miRNA suppresses the translation of the TF messenger RNA. FBLs are potential sources of instability in a gene regulatory network. Positive FBLs can give rise to switching behaviors while negative FBLs can generate periodic oscillations. This chapter presents documented examples of FBLs and their relevance to stem cell renewal and differentiation in gliomas. Feed-forward loops (FFLs) are only discussed briefly because they do not affect network stability unless they are members of cycles. A primer on qualitative network stability analysis is given and then used to demonstrate the network destabilizing role of FBLs. Steps in model formulation and computer simulations are illustrated using the miR-17-92/Myc/E2F network as an example. This example possesses both negative and positive FBLs.


Subject(s)
Gene Regulatory Networks/genetics , MicroRNAs/metabolism , Models, Genetic , Neoplasms/genetics , Transcription Factors/metabolism , Animals , Feedback, Physiological , Humans , MicroRNAs/genetics , Transcription Factors/genetics
5.
Curr Drug Discov Technol ; 10(2): 125-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23237675

ABSTRACT

With increasing knowledge of cellular networks of gene and molecular interactions, and their alterations in GBM (glioblastoma multiforme), it is now possible to apply methods of Network Pharmacology (NP) to predict candidate drug targets for this malignant brain tumor. NP requires the development of mathematical methods for network stability and perturbation analysis to identify sensitive and druggable network components, as well as computational platforms to carry out in silico simulations of therapeutic interventions. This review focuses on the three most frequently deregulated GBM pathways involving membrane receptor tyrosine kinases, p53, and Rb. Structural features of these networks that may confound targeted therapies are discussed.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Retinoblastoma Protein/metabolism , Systems Biology , Tumor Suppressor Protein p53/metabolism
6.
Biophys J ; 101(9): 2082-91, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22067145

ABSTRACT

A kinetic model of a molecular control system for the cellular decision to proliferate or differentiate is formulated and analyzed for the purpose of understanding how the system can break down in cancer cells. The proposed core of this control system is composed of the transcription factors Myc and p53. The network of interactions between these factors involves negative and positive feedback loops that are linked to pathways involved in differentiation, cell cycle, and apoptosis. Understanding the dynamics of the Myc-p53 control system is aided by the postulate that there exists a cancer zone defined as a range of oncogenic Myc activities where the probability of initiating cancer is high. We propose that an essential role of p53 is to prevent the system from entering or staying too long in the cancer zone by downregulating Myc or, when Myc activity somehow becomes too high, by inducing apoptosis, cell cycle arrest, or differentiation. Kinetic modeling illustrates how deletions or aberrations in PTEN, MDM2, and ARF (genes implicated in various cancers, including glioma) affect the Myc-p53 control system. In addition, computer simulations demonstrate how this control system generates different cellular phenotypes characterized by rates of cellular differentiation and proliferation.


Subject(s)
Cell Differentiation , Models, Biological , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cell Proliferation , Feedback, Physiological , Gene Deletion , Humans , Kinetics , Mice , PTEN Phosphohydrolase/metabolism , Phenotype , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Time Factors
7.
PLoS One ; 6(5): e19544, 2011.
Article in English | MEDLINE | ID: mdl-21637752

ABSTRACT

BACKGROUND: Sarcoidosis is a polygenic disease with diverse phenotypic presentations characterized by an abnormal antigen-mediated Th1 type immune response. At present, progress towards understanding sarcoidosis disease mechanisms and the development of novel treatments is limited by constraints attendant to conducting human research in a rare disease in the absence of relevant animal models. We sought to develop a computational model to enhance our understanding of the pathological mechanisms of and predict potential treatments of sarcoidosis. METHODOLOGY/RESULTS: Based upon the literature, we developed a computational model of known interactions between essential immune cells (antigen-presenting macrophages, effector and regulatory T cells) and cytokine mediators (IL-2, TNFα, IFNγ) of granulomatous inflammation during sarcoidosis. The dynamics of these interactions are described by a set of ordinary differential equations. The model predicts bistable switching behavior which is consistent with normal (self-limited) and "sarcoidosis-like" (sustained) activation of the inflammatory components of the system following a single antigen challenge. By perturbing the influence of model components using inhibitors of the cytokine mediators, distinct clinically relevant disease phenotypes were represented. Finally, the model was shown to be useful for pre-clinical testing of therapies based upon molecular targets and dose-effect relationships. CONCLUSIONS/SIGNIFICANCE: Our work illustrates a dynamic computer simulation of granulomatous inflammation scenarios that is useful for the investigation of disease mechanisms and for pre-clinical therapeutic testing. In lieu of relevant in vitro or animal surrogates, our model may provide for the screening of potential therapies for specific sarcoidosis disease phenotypes in advance of expensive clinical trials.


Subject(s)
Computer Simulation , Models, Immunological , Sarcoidosis/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Antigens/immunology , Cell Communication/drug effects , Drug Therapy, Combination , Granuloma/immunology , Granuloma/pathology , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Phenotype , Sarcoidosis/drug therapy , Sarcoidosis/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/immunology
8.
Blood ; 114(18): 3872-9, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19717645

ABSTRACT

Chromosomal abnormalities, immunoglobulin heavy chain variable-region (IGHV) gene mutation status, and zeta-associated protein 70 (ZAP-70) expression levels have independent prognostic relevance in chronic lymphocytic leukemia (CLL); however, their concordance is variable. Because deregulation of microRNAs has been linked to disease initiation and progression in CLL, we studied the value of the microRNAs as a signature for CLL patients with specific chromosomal abnormalities. We identified 32 microRNAs able to discriminate the 11q deletion, 17p deletion, trisomy 12, 13q deletion, and normal karyotype cytogenetic subgroups. The expression values of 9 among the 32 microRNAs (miR-151-3p, miR-34a, miR-29c, miR-29b, miR-155, miR-148a, miR-146a, miR-146b5p, and miR-640) were correlated with gene expression data from the same samples to assess their biologic impact on CLL. In this study we also found that IGHV unmutated, high expression of ZAP-70 protein, and low expression of the miR-223, miR-29c, miR-29b, and miR-181 family were strongly associated with disease progression in CLL cases harboring 17p deletion, whereas in those harboring trisomy 12 only high expression of the miR-181a, among the analyzed parameters, suggested more aggressive disease. Thus, the use of the microRNA-based classifications may yield clinically useful biomarkers of tumor behavior in CLL.


Subject(s)
Biomarkers, Tumor/biosynthesis , Chromosome Deletion , Chromosomes, Human , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MicroRNAs/biosynthesis , RNA, Neoplasm/biosynthesis , Biomarkers, Tumor/genetics , Female , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , MicroRNAs/genetics , RNA, Neoplasm/genetics , Trisomy , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
9.
PLoS One ; 4(4): e5262, 2009.
Article in English | MEDLINE | ID: mdl-19370157

ABSTRACT

BACKGROUND: During normal physical activities cartilage experiences dynamic compressive forces that are essential to maintain cartilage integrity. However, at non-physiologic levels these signals can induce inflammation and initiate cartilage destruction. Here, by examining the pro-inflammatory signaling networks, we developed a mathematical model to show the magnitude-dependent regulation of chondrocytic responses by compressive forces. METHODOLOGY/PRINCIPAL FINDINGS: Chondrocytic cells grown in 3-D scaffolds were subjected to various magnitudes of dynamic compressive strain (DCS), and the regulation of pro-inflammatory gene expression via activation of nuclear factor-kappa B (NF-kappaB) signaling cascade examined. Experimental evidences provide the existence of a threshold in the magnitude of DCS that regulates the mRNA expression of nitric oxide synthase (NOS2), an inducible pro-inflammatory enzyme. Interestingly, below this threshold, DCS inhibits the interleukin-1beta (IL-1beta)-induced pro-inflammatory gene expression, with the degree of suppression depending on the magnitude of DCS. This suppression of NOS2 by DCS correlates with the attenuation of the NF-kappaB signaling pathway as measured by IL-1beta-induced phosphorylation of the inhibitor of kappa B (IkappaB)-alpha, degradation of IkappaB-alpha and IkappaB-beta, and subsequent nuclear translocation of NF-kappaB p65. A mathematical model developed to understand the complex dynamics of the system predicts two thresholds in the magnitudes of DCS, one for the inhibition of IL-1beta-induced expression of NOS2 by DCS at low magnitudes, and second for the DCS-induced expression of NOS2 at higher magnitudes. CONCLUSIONS/SIGNIFICANCE: Experimental and computational results indicate that biomechanical signals suppress and induce inflammation at critical thresholds through activation/suppression of the NF-kappaB signaling pathway. These thresholds arise due to the bistable behavior of the networks originating from the positive feedback loop between NF-kappaB and its target genes. These findings lay initial groundwork for the identification of the thresholds in physical activities that can differentiate its favorable actions from its unfavorable consequences on joints.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Inflammation/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Stress, Mechanical , Biomechanical Phenomena , Cartilage/cytology , Cartilage/physiopathology , Cell Line , Gene Expression Regulation , Humans , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation/physiopathology , Interleukin-1beta/metabolism , Models, Biological , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Nitric Oxide Synthase/genetics , Protein Transport , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
PLoS One ; 4(2): e4407, 2009.
Article in English | MEDLINE | ID: mdl-19197384

ABSTRACT

Intracellular protein levels of p53 and MDM2 have been shown to oscillate in response to ionizing radiation (IR), but the physiological significance of these oscillations remains unclear. The p53-MDM2 negative feedback loop -- the putative cause of the oscillations -- is embedded in a network involving a mutual antagonism (or positive feedback loop) between p53 and AKT. We have shown earlier that this p53-AKT network predicts an all-or-none switching behavior between a pro-survival cellular state (low p53 and high AKT levels) and a pro-apoptotic state (high p53 and low AKT levels). Here, we show that upon exposure to IR, the p53-AKT network can also reproduce the experimentally observed p53 and MDM2 oscillations. The present work is based on the hypothesis that the physiological significance of the experimentally observed oscillations could be found in their role in regulating the switching behavior of the p53-AKT network between pro-survival and pro-apoptotic states. It is shown here that these oscillations are associated with a significant decrease in the threshold level of IR at which switching from a pro-survival to a pro-apoptotic state occurs. Moreover, oscillations in p53 protein levels induce higher levels of expression of p53-target genes compared to non-oscillatory p53, and thus influence cell-fate decisions between cell cycle arrest/DNA damage repair versus apoptosis.


Subject(s)
Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Cell Death/radiation effects , Cell Survival/radiation effects , Gene Expression Regulation/radiation effects , Gene Regulatory Networks/radiation effects , Humans , Kinetics , Organ Specificity/radiation effects , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation, Ionizing , Transcription, Genetic/radiation effects
11.
Proc Natl Acad Sci U S A ; 105(50): 19678-83, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19066217

ABSTRACT

The transcription factors E2F and Myc participate in the control of cell proliferation and apoptosis, and can act as oncogenes or tumor suppressors depending on their levels of expression. Positive feedback loops in the regulation of these factors are predicted-and recently shown experimentally-to lead to bistability, which is a phenomenon characterized by the existence of low and high protein levels ("off" and "on" levels, respectively), with sharp transitions between levels being inducible by, for example, changes in growth factor concentrations. E2F and Myc are inhibited at the posttranscriptional step by members of a cluster of microRNAs (miRs) called miR-17-92. In return, E2F and Myc induce the transcription of miR-17-92, thus forming a negative feedback loop in the interaction network. The consequences of the coupling between the E2F/Myc positive feedback loops and the E2F/Myc/miR-17-92 negative feedback loop are analyzed using a mathematical model. The model predicts that miR-17-92 plays a critical role in regulating the position of the off-on switch in E2F/Myc protein levels, and in determining the on levels of these proteins. The model also predicts large-amplitude protein oscillations that coexist with the off steady state levels. Using the concept and model prediction of a "cancer zone," the oncogenic and tumor suppressor properties of miR-17-92 is demonstrated to parallel the same properties of E2F and Myc.


Subject(s)
E2F Transcription Factors/metabolism , Gene Regulatory Networks , MicroRNAs/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/metabolism , E2F Transcription Factors/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , MicroRNAs/genetics , Models, Biological , Neoplasms/genetics , Oncogene Proteins , Oncogenes , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Proteins/genetics
12.
PLoS One ; 3(11): e3694, 2008.
Article in English | MEDLINE | ID: mdl-19002258

ABSTRACT

BACKGROUND: MicroRNAs (miRNA) are small non-coding RNAs that regulate translation of mRNA and protein. Loss or enhanced expression of miRNAs is associated with several diseases, including cancer. However, the identification of circulating miRNA in healthy donors is not well characterized. Microvesicles, also known as exosomes or microparticles, circulate in the peripheral blood and can stimulate cellular signaling. In this study, we hypothesized that under normal healthy conditions, microvesicles contain miRNAs, contributing to biological homeostasis. METHODOLOGY/PRINCIPAL FINDINGS: Microvesicles were isolated from the plasma of normal healthy individuals. RNA was isolated from both the microvesicles and matched mononuclear cells and profiled for 420 known mature miRNAs by real-time PCR. Hierarchical clustering of the data sets indicated significant differences in miRNA expression between peripheral blood mononuclear cells (PBMC) and plasma microvesicles. We observed 71 miRNAs co-expressed between microvesicles and PBMC. Notably, we found 33 and 4 significantly differentially expressed miRNAs in the plasma microvesicles and mononuclear cells, respectively. Prediction of the gene targets and associated biological pathways regulated by the detected miRNAs was performed. The majority of the miRNAs expressed in the microvesicles from the blood were predicted to regulate cellular differentiation of blood cells and metabolic pathways. Interestingly, a select few miRNAs were also predicted to be important modulators of immune function. CONCLUSIONS: This study is the first to identify and define miRNA expression in circulating plasma microvesicles of normal subjects. The data generated from this study provides a basis for future studies to determine the predictive role of peripheral blood miRNA signatures in human disease and will enable the definition of the biological processes regulated by these miRNA.


Subject(s)
Exosomes/metabolism , MicroRNAs/blood , Adult , Age Factors , Female , Gender Identity , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/metabolism , Male , MicroRNAs/metabolism , Middle Aged
14.
Theor Biol Med Model ; 4: 30, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17692115

ABSTRACT

BACKGROUND: The monolayer of endothelial cells (ECs) lining the inner wall of blood vessels deteriorates as a person ages due to a complex interplay of a variety of causes including cell death arising from shear stress of blood flow and cellular oxidative stress, cellular senescence, and decreased rate of replacement of dead ECs by progenitor stem cells. RESULTS: A continuum mathematical model is developed to describe the dynamics of large EC populations of the endothelium using a system of differential equations for the number densities of cells of different generations starting from endothelial progenitors to senescent cells, as well as the densities of dead cells and the holes created upon clearing dead cells. Aging of cells is manifested in three ways, namely, losing the ability to divide when the Hayflick limit of 50 generations is reached, decreasing replication rate parameters and increasing death rate parameters as cells divide; due to the dependence of these rate parameters on cell generation, the model predicts a narrow distribution of cell densities peaking at a particular cell generation. As the chronological age of a person advances, the peak of the distribution - corresponding to the age of the endothelium - moves towards senescence correspondingly. However, computer simulations also demonstrate that sustained and enhanced stem cell homing can halt the aging process of the endothelium by maintaining a stationary cell density distribution that peaks well before the Hayflick limit. The healing rates of damaged endothelia for young, middle-aged, and old persons are compared and are found to be particularly sensitive to the stem cell homing parameter. CONCLUSION: The proposed model describes the aging of the endothelium as being driven by cellular senescence, with a rate that does not necessarily correspond to the chronological aging of a person. It is shown that the age of the endothelium depends sensitively on the homing rates of EC progenitor cells.


Subject(s)
Cellular Senescence/physiology , Endothelial Cells/physiology , Models, Biological , Computer Simulation
15.
Biophys J ; 91(3): 857-65, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16648169

ABSTRACT

The tumor suppressor protein, p53, and the oncoprotein, Akt, are involved in a cross talk that could be at the core of a cell's control machinery for switching between survival and death. This cross talk is a combination of reciprocally antagonistic pathways emanating from p53 and Akt, and also involves another tumor suppressor gene, PTEN, and another oncogene, Mdm2; such a connected network of cancer-relevant genes must be significant and demands a critical study. The p53-Akt network is shown in this report to possess the potential to exhibit bistability, a phenomenon in which two stable steady states of the system coexist for a fixed set of control parameter values. A hierarchy of qualitative networks and abstract kinetic models are analyzed and simulated on a computer to demonstrate the robustness of the bistable behavior, which, as argued in this study, is a likely candidate mechanism for a cellular survival-death switch. The analysis applies to cells that are neither p53-null nor Akt-null. The models presented here offer experimental predictions on the identity of control parameters of apoptotic thresholds and on network perturbations (including DNA damage and Akt inhibition) that are sufficient to generate switching between pro-survival and pro-death cellular states.


Subject(s)
Genes, Tumor Suppressor , Proto-Oncogene Proteins c-akt/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis , Biophysics/methods , Cell Death , Cell Survival , Computer Simulation , DNA Damage , Humans , Kinetics , Models, Biological , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
17.
Cell Cycle ; 2(6): 538-44, 2003.
Article in English | MEDLINE | ID: mdl-14504470

ABSTRACT

This paper proposes an integration and modular organization of the complex regulatory networks involved in the mammalian cell cycle, apoptosis, and related intracellular signaling cascades. A common node linking the cell cycle and apoptosis permits the possibility of coordinate control between the initiation of these two cellular processes. From this node, pathways emanate that lead to the activation of cyclin-dependent kinases (in the cell cycle) and caspases (in apoptosis). Computer simulations are carried out to demonstrate that the proposed network architecture and certain module-module interactions can account for the experimentally observed sequence of cellular events (quiescence, cell cycle, and apoptosis) as the transcriptional activities of E2F-1 and c-Myc are increased. Despite the lack of quantitative kinetic data on most of the pathways, it is demonstrated that there can be meaningful conclusions regarding system stability that arise from the topology of the network. It is shown that only cycles in the network graph determine stability. Thus, several positive and negative feedback loops are identified from a literature review of the major pathways involved in the initiation of the cell cycle and of apoptosis.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Computer Simulation , Signal Transduction/physiology , Animals , Cell Cycle Proteins/metabolism , Enzyme Activation
18.
Chaos ; 11(1): 269-276, 2001 Mar.
Article in English | MEDLINE | ID: mdl-12779460

ABSTRACT

The essential genes, proteins and associated regulatory networks involved in the entry into the mammalian cell cycle are identified, from activation of growth-factor receptors to intracellular signal transduction pathways that impinge on the cell cycle machinery and ultimately on the initiation of DNA replication. Signaling pathways mediated by the oncoproteins Ras and Myc induce the activation of cyclin-dependent kinases CDK4 and CDK2, and the assembly and firing of pre-replication complexes require a collaboration among E2F, CDK2, and Cdc7 kinase. A proposed core mechanism of the restriction point, the major checkpoint prior to commitment to DNA synthesis, involves cyclin E/CDK2, the phosphatase Cdc25A, and the CDK inhibitor p27Kip1. (c) 2001 American Institute of Physics.

SELECTION OF CITATIONS
SEARCH DETAIL
...