Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
EMBO J ; 43(7): 1187-1213, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38383863

ABSTRACT

Histone modifications commonly integrate environmental cues with cellular metabolic outputs by affecting gene expression. However, chromatin modifications such as acetylation do not always correlate with transcription, pointing towards an alternative role of histone modifications in cellular metabolism. Using an approach that integrates mass spectrometry-based histone modification mapping and metabolomics with stable isotope tracers, we demonstrate that elevated lipids in acetyltransferase-depleted hepatocytes result from carbon atoms derived from deacetylation of hyperacetylated histone H4 flowing towards fatty acids. Consistently, enhanced lipid synthesis in acetyltransferase-depleted hepatocytes is dependent on histone deacetylases and acetyl-CoA synthetase ACSS2, but not on the substrate specificity of the acetyltransferases. Furthermore, we show that during diet-induced lipid synthesis the levels of hyperacetylated histone H4 decrease in hepatocytes and in mouse liver. In addition, overexpression of acetyltransferases can reverse diet-induced lipogenesis by blocking lipid droplet accumulation and maintaining the levels of hyperacetylated histone H4. Overall, these findings highlight hyperacetylated histones as a metabolite reservoir that can directly contribute carbon to lipid synthesis, constituting a novel function of chromatin in cellular metabolism.


Subject(s)
Carbon , Histones , Animals , Mice , Histones/metabolism , Carbon/metabolism , Lipogenesis , Chromatin , Acetyltransferases/metabolism , Lipids , Acetylation , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism
2.
J Med Virol ; 95(12): e29264, 2023 12.
Article in English | MEDLINE | ID: mdl-38054553

ABSTRACT

The Octamer-binding transcription factor-4 (Oct4) is upregulated in different malignancies, yet a paradigm for mechanisms of Oct4 post-embryonic re-expression is inadequately understood. In cervical cancer, Oct4 expression is higher in human papillomavirus (HPV)-related than HPV-unrelated cervical cancers and this upregulation correlates with the expression of the E7 oncogene. We have reported that E7 affects the Oct4-transcriptional output and Oct4-related phenotypes in cervical cancer, however, the underlying mechanism remains elusive. Here, we characterize the Oct4-protein interactions in cervical cancer cells via computational analyses and Mass Spectrometry and reveal that Methyl-binding proteins (MBD2 and MBD3), are determinants of Oct4-driven transcription. E7 triggers MBD2 downregulation and TET1 upregulation, thereby disrupting the methylation status of the Oct4 gene. This coincides with an increase in the total DNA hydroxymethylation leading to the re-expression of Oct4 in cervical cancer and likely affecting broader transcriptional patterns. Our findings reveal a previously unreported mechanism by which the E7 oncogene can regulate Oct4 re-expression and global transcriptional patterns by increasing DNA hydroxymethylation and lowering the barrier to cellular plasticity during carcinogenesis.


Subject(s)
Octamer Transcription Factor-3 , Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , DNA , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mixed Function Oxygenases , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Proto-Oncogene Proteins , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Octamer Transcription Factor-3/genetics
3.
BMC Biol ; 20(1): 22, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35057804

ABSTRACT

BACKGROUND: Epigenetic regulation relies on the activity of enzymes that use sentinel metabolites as cofactors to modify DNA or histone proteins. Thus, fluctuations in cellular metabolite levels have been reported to affect chromatin modifications. However, whether epigenetic modifiers also affect the levels of these metabolites and thereby impinge on downstream metabolic pathways remains largely unknown. Here, we tested this notion by investigating the function of N-alpha-acetyltransferase 40 (NAA40), the enzyme responsible for N-terminal acetylation of histones H2A and H4, which has been previously implicated with metabolic-associated conditions such as age-dependent hepatic steatosis and calorie-restriction-mediated longevity. RESULTS: Using metabolomic and lipidomic approaches, we found that depletion of NAA40 in murine hepatocytes leads to significant increase in intracellular acetyl-CoA levels, which associates with enhanced lipid synthesis demonstrated by upregulation in de novo lipogenesis genes as well as increased levels of diglycerides and triglycerides. Consistently, the increase in these lipid species coincide with the accumulation of cytoplasmic lipid droplets and impaired insulin signalling indicated by decreased glucose uptake. However, the effect of NAA40 on lipid droplet formation is independent of insulin. In addition, the induction in lipid synthesis is replicated in vivo in the Drosophila melanogaster larval fat body. Finally, supporting our results, we find a strong association of NAA40 expression with insulin sensitivity in obese patients. CONCLUSIONS: Overall, our findings demonstrate that NAA40 affects the levels of cellular acetyl-CoA, thereby impacting lipid synthesis and insulin signalling. This study reveals a novel path through which histone-modifying enzymes influence cellular metabolism with potential implications in metabolic disorders.


Subject(s)
Histone Acetyltransferases , Histones , N-Terminal Acetyltransferase D/metabolism , Acetyl Coenzyme A/metabolism , Animals , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Insulin/metabolism , Lipids , Lipogenesis , Mice
4.
PLoS Pathog ; 16(4): e1008468, 2020 04.
Article in English | MEDLINE | ID: mdl-32298395

ABSTRACT

Octamer binding transcription factor-4 (Oct4), is highly expressed in stem cells and has indispensable roles in pluripotency and cellular reprogramming. In contrast to other factors used for cellular reprogramming, a role for Oct4 outside embryonic stem cells has been elusive and highly controversial. Emerging evidence implicates Oct4 in the carcinogenic process, but the mechanism through which Oct4 may be functioning in cancers is not fully appreciated. Here, we provide evidence that Oct4 is expressed in human cervical cancer and this expression correlates with the presence of the human papillomavirus (HPV) oncogenes E6 and E7. Surprisingly, the viral oncogenes can complement exogenously provided Oct4 in reprogramming assays, providing functional validation for their ability to activate Oct4 transcription in Mouse Embryonic Fibroblasts (MEFs). To interrogate potential roles of Oct4 in cervical cancers we knocked-down Oct4 in HPV(+) (HeLa & CaSki) and HPV(-) (C33A) cervical cancer cell lines and found that Oct4 knockdown attenuated clonogenesis, only in the HPV(+) cells. More unexpectedly, cell proliferation and migration, were differentially affected in HPV(+) and HPV(-) cell lines. We provide evidence that Oct4 interacts with HPV E7 specifically at the CR3 region of the E7 protein and that introduction of the HPV oncogenes in C33A cells and human immortalised keratinocytes generates Oct4-associated transcriptional and phenotypic patterns, which mimic those seen in HPV(+) cells. We propose that a physical interaction of Oct4 with E7 regulates its activity in HPV(+) cervical cancers in a manner not seen in other cancer types.


Subject(s)
Octamer Transcription Factor-3/metabolism , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , HeLa Cells , Humans , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Oncogene Proteins, Viral/metabolism , Oncogenes/physiology , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Repressor Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
5.
Int J Mol Sci ; 23(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36430521

ABSTRACT

PURPOSE: To investigate the anti-cancer, chemosensitizing and/or immunomodulating effects of decitabine (DAC) to be used as a potential therapeutic agent for the treatment of cervical cancer (CC). METHODS: Cervical cancer cell lines were treated with low doses of DAC treatment used as a single agent or in combination with chemotherapy. End-point in vitro assays were developed as indicators of the anti-cancer and/or immunomodulating effects of DAC treatment in CC cells. These assays include cell viability, cell cycle analysis, apoptosis, induction of a viral-mimicry response pathway, expression of MHC-class I and PD-L1 and chemosensitivity. RESULTS: High and low doses of DAC treatment induced reduction in cell viability in HeLa (HPV18+), CaSki (HPV16+) and C33A (HPV-) cells. Specifically, a time-dependent reduction in cell viability of HeLa and CaSki cells was observed accompanied by robust cell cycle arrest at G2/M phase and alterations in the cell cycle distribution. Decrease in cell viability was also observed in a non-transformed immortal keratinocyte (HaCat) suggesting a non-cancer specific target effect. DAC treatment also triggered a viral mimicry response through long-term induction of cytoplasmic double-stranded RNA (dsRNA) and activation of downstream IFN-related genes in both HPV+ and HPV- cells. In addition, DAC treatment increased the number of CC cells expressing MHC-class I and PD-L1. Furthermore, DAC significantly increased the proportion of early and late apoptotic CC cells quantified using FACS. Our combination treatments showed that low dose DAC treatment sensitizes cells to chemotherapy. CONCLUSIONS: Low doses of DAC treatment promotes robust induction of a viral mimicry response, immunomodulating and chemosensitizing effects in CC, indicating its promising therapeutic role in CC in vitro.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/drug therapy , Decitabine/pharmacology , B7-H1 Antigen , HeLa Cells
6.
Nature ; 460(7259): 1136-9, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19668188

ABSTRACT

The mechanisms involved in the reprogramming of differentiated cells into induced pluripotent stem (iPS) cells by the three transcription factors Oct4 (also known as Pou5f1), Klf4 and Sox2 remain poorly understood. The Ink4/Arf locus comprises the Cdkn2a-Cdkn2b genes encoding three potent tumour suppressors, namely p16(Ink4a), p19(Arf) and p15(Ink4b), which are basally expressed in differentiated cells and upregulated by aberrant mitogenic signals. Here we show that the locus is completely silenced in iPS cells, as well as in embryonic stem (ES) cells, acquiring the epigenetic marks of a bivalent chromatin domain, and retaining the ability to be reactivated after differentiation. Cell culture conditions during reprogramming enhance the expression of the Ink4/Arf locus, further highlighting the importance of silencing the locus to allow proliferation and reprogramming. Indeed, the three factors together repress the Ink4/Arf locus soon after their expression and concomitant with the appearance of the first molecular markers of 'stemness'. This downregulation also occurs in cells carrying the oncoprotein large-T, which functionally inactivates the pathways regulated by the Ink4/Arf locus, thus indicating that the silencing of the locus is intrinsic to reprogramming and not the result of a selective process. Genetic inhibition of the Ink4/Arf locus has a profound positive effect on the efficiency of iPS cell generation, increasing both the kinetics of reprogramming and the number of emerging iPS cell colonies. In murine cells, Arf, rather than Ink4a, is the main barrier to reprogramming by activation of p53 (encoded by Trp53) and p21 (encoded by Cdkn1a); whereas, in human fibroblasts, INK4a is more important than ARF. Furthermore, organismal ageing upregulates the Ink4/Arf locus and, accordingly, reprogramming is less efficient in cells from old organisms, but this defect can be rescued by inhibiting the locus with a short hairpin RNA. All together, we conclude that the silencing of Ink4/Arf locus is rate-limiting for reprogramming, and its transient inhibition may significantly improve the generation of iPS cells.


Subject(s)
Cellular Reprogramming/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Aging/physiology , Animals , Cell Count , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Silencing , Humans , Keratinocytes , Kinetics , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL
7.
Nature ; 460(7259): 1149-53, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19668189

ABSTRACT

The reprogramming of differentiated cells to pluripotent cells (induced pluripotent stem (iPS) cells) is known to be an inefficient process. We recently reported that cells with short telomeres cannot be reprogrammed to iPS cells despite their normal proliferation rates, probably reflecting the existence of 'reprogramming barriers' that abort the reprogramming of cells with uncapped telomeres. Here we show that p53 (also known as Trp53 in mice and TP53 in humans) is critically involved in preventing the reprogramming of cells carrying various types of DNA damage, including short telomeres, DNA repair deficiencies, or exogenously inflicted DNA damage. Reprogramming in the presence of pre-existing, but tolerated, DNA damage is aborted by the activation of a DNA damage response and p53-dependent apoptosis. Abrogation of p53 allows efficient reprogramming in the face of DNA damage and the generation of iPS cells carrying persistent DNA damage and chromosomal aberrations. These observations indicate that during reprogramming cells increase their intolerance to different types of DNA damage and that p53 is critical in preventing the generation of human and mouse pluripotent cells from suboptimal parental cells.


Subject(s)
Cellular Reprogramming/physiology , DNA Damage/physiology , Genomic Instability/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cells, Cultured , Chromosome Aberrations , DNA Damage/genetics , DNA Repair , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genomic Instability/genetics , Humans , Male , Mice , Telomere/genetics , Telomere/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
8.
Cell Rep ; 43(3): 113859, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38421873

ABSTRACT

Oct4 is a pioneer transcription factor regulating pluripotency. However, it is not well known whether Oct4 has an impact on epidermal cells. We generated OCT4 knockout clonal cell lines using immortalized human skin keratinocytes to identify a functional role for the protein. Here, we report that Oct4-deficient cells transitioned into a mesenchymal-like phenotype with enlarged size and shape, exhibited accelerated migratory behavior, decreased adhesion, and appeared arrested at the G2/M cell cycle checkpoint. Oct4 absence had a profound impact on cortical actin organization, with loss of microfilaments from the cell membrane, increased puncta deposition in the cytoplasm, and stress fiber formation. E-cadherin, ß-catenin, and ZO1 were almost absent from cell-cell contacts, while fibronectin deposition was markedly increased in the extracellular matrix (ECM). Mapping of the transcriptional and chromatin profiles of Oct4-deficient cells revealed that Oct4 controls the levels of cytoskeletal, ECM, and differentiation-related genes, whereas epithelial identity is preserved through transcriptional and non-transcriptional mechanisms.


Subject(s)
Cadherins , Keratinocytes , Humans , Cadherins/metabolism , Keratinocytes/metabolism , Cytoskeleton/metabolism , Actins/metabolism , beta Catenin/metabolism , Skin/metabolism , Cell Adhesion/physiology
9.
STAR Protoc ; 4(1): 101994, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36607810

ABSTRACT

Here, we present a protocol to create an in vivo lineage-tracing mouse model for mouse-papillomavirus-type 1 (MmuPV1)-infected cells. We describe the steps to generate and deliver the MmuPV1 lox-Cre-lox plasmid for the infection of mice, followed by skin tissue extraction and processing. We then detail how to use flow cytometry to trace, quantify, and analyze MmuPV1-harboring cells and their progeny. This model is suitable to investigate the early effects of papillomavirus on the target cells. For complete details on the use and execution of this protocol, please refer to Yilmaz et al. (2022).1.


Subject(s)
Flow Cytometry , Papillomaviridae , Papillomavirus Infections , Animals , Mice
10.
Viruses ; 15(1)2023 01 16.
Article in English | MEDLINE | ID: mdl-36680299

ABSTRACT

Scientific progress in understanding, preventing, treating, and managing viral infections and associated diseases exemplifies the extent to which research on small DNA tumor viruses has impacted human health [...].


Subject(s)
DNA Viruses , Virus Diseases , Humans , DNA Viruses/genetics , DNA Tumor Viruses
11.
Elife ; 112022 05 09.
Article in English | MEDLINE | ID: mdl-35533001

ABSTRACT

Human papillomaviruses are DNA viruses that ubiquitously infect humans and have been associated with hyperproliferative lesions. The recently discovered mouse specific papillomavirus (MmuPV1) provides the opportunity to study papillomavirus infections in vivo in the context of a common laboratory mouse model (Mus musculus). To date, a major challenge in the field has been the lack of tools to identify, observe, and characterize individually the papillomavirus hosting cells and also trace the progeny of these cells over time. Here, we present the successful generation of an in vivo lineage-tracing model of MmuPV1-harboring cells and their progeny by means of genetic reporter activation. Following the validation of the system both in vitro and in vivo, we used it to provide a proof-of-concept of its utility. Using flow-cytometry analysis, we observed increased proliferation dynamics and decreased MHC-I cell surface expression in MmuPV1-treated tissues which could have implications in tissue regenerative capacity and ability to clear the virus. This model is a novel tool to study the biology of the MmuPV1 host-pathogen interactions.


Subject(s)
Papillomaviridae , Papillomavirus Infections , Animals , Disease Models, Animal , Mice , Papillomaviridae/genetics
12.
Nanotheranostics ; 5(1): 113-124, 2021.
Article in English | MEDLINE | ID: mdl-33391978

ABSTRACT

Treatment of breast cancer underwent extensive progress in recent years with molecularly targeted therapies. However, non-specific pharmaceutical approaches (chemotherapy) persist, inducing severe side-effects. Phytochemicals provide a promising alternative for breast cancer prevention and treatment. Specifically, resveratrol (res) is a plant-derived polyphenolic phytoalexin with potent biological activity but displays poor water solubility, limiting its clinical use. Here we have developed a strategy for delivering res using a newly synthesized nano-carrier with the potential for both diagnosis and treatment. Methods: Res-loaded nanoparticles were synthesized by the emulsion method using Pluronic F127 block copolymer and Vitamin E-TPGS. Nanoparticle characterization was performed by SEM and tunable resistive pulse sensing. Encapsulation Efficiency (EE%) and Drug Loading (DL%) content were determined by analysis of the supernatant during synthesis. Nanoparticle uptake kinetics in breast cancer cell lines MCF-7 and MDA-MB-231 as well as in MCF-10A breast epithelial cells were evaluated by flow cytometry and the effects of res on cell viability via MTT assay. Results: Res-loaded nanoparticles with spherical shape and a dominant size of 179±22 nm were produced. Res was loaded with high EE of 73±0.9% and DL content of 6.2±0.1%. Flow cytometry revealed higher uptake efficiency in breast cancer cells compared to the control. An MTT assay showed that res-loaded nanoparticles reduced the viability of breast cancer cells with no effect on the control cells. Conclusions: These results demonstrate that the newly synthesized nanoparticle is a good model for the encapsulation of hydrophobic drugs. Additionally, the nanoparticle delivers a natural compound and is highly effective and selective against breast cancer cells rendering this type of nanoparticle an excellent candidate for diagnosis and therapy of difficult to treat mammary malignancies.


Subject(s)
Breast Neoplasms/drug therapy , Drug Carriers , Micelles , Resveratrol/therapeutic use , Antineoplastic Agents/therapeutic use , Female , Humans , MCF-7 Cells
13.
Comput Methods Programs Biomed ; 185: 105165, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31710982

ABSTRACT

BACKGROUND & OBJECTIVE: In this work, we focus on estimating the parameters of the Gompertz model in order to predict tumor growth. The estimation is based on measurements from mice skin tumors of de novo carcinogenesis. The main objective is to compare the Maximum Likelihood estimator with the best performance from our previous work with the Non-linear Least Squares estimator which is commonly used in the literature to estimate the growth parameters of the Gompertz model. METHODS: To describe tumor growth, we propose a stochastic model which is based on the Gompertz growth function. The principle of Maximum Likelihood is used to estimate both the growth rate and the carrying capacity of the Gompertz function, along with the characteristics of the additive Gaussian process and measurement noise. Moreover, we examine whether a Maximum A Posteriori estimator is able to utilize any available prior knowledge in order to improve the predictions. RESULTS: Experimental data from a total of 24 tumors in 8 mice (3 tumors each) were used to study the performance of the proposed methods with respect to prediction accuracy. Our results show that the Maximum Likelihood estimator is able to provide, in most cases, more accurate predictions. Moreover, the Maximum A Posteriori estimator has the potential to correct potentially non-realistic estimates for the carrying capacity at early growth stages. CONCLUSION: In most cases, the Maximum Likelihood estimator is able to provide more reliable predictions for the tumor's growth on individual test subjects. The Maximum A Posteriori estimator, it has the potential to improve the prediction when the available experimental data do not provide adequate information by utilizing prior knowledge about the unknown parameters.


Subject(s)
Skin Neoplasms/pathology , Animals , Likelihood Functions , Mice , Mice, Transgenic
14.
PLoS One ; 13(4): e0196604, 2018.
Article in English | MEDLINE | ID: mdl-29698462

ABSTRACT

High-risk human papillomaviruses (HPVs) have been shown in vitro to impinge on telomere homeostasis in a number of ways. However, the in vivo interaction of viruses with the telomere homeostasis apparatus has not been previously explored. Since E6 and E7 are the main viral oncogenes and key for viral replication, we have explored here the short-term phenotypes of the genes in the context of defective telomere homeostasis. We examined the short-term phenotypes of E6 and E7 in a context where the Terc component of the telomerase holoenzyme was knocked out. We determined that Terc was dispensable for most oncogene-mediated phenotypes. Surprisingly, E7-mediated reduction of label retaining cells was found to be in part dependent on the presence of Terc. Under the conditions examined here, there appears to be no compelling evidence Terc is required for most short-term viral oncogene mediated phenotypes. Further studies will elucidate its role in longer-term phenotypes.


Subject(s)
Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , Telomerase/metabolism , Animals , Cell Nucleus/metabolism , Genotype , In Situ Hybridization, Fluorescence , Keratin-15/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Phenotype , Repressor Proteins/metabolism , Telomerase/genetics , Telomere/genetics , Telomere/metabolism
15.
Data Brief ; 20: 1602-1606, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30263912

ABSTRACT

We have performed whole transcriptome sequencing of 5-FU resistant and 5-FU sensitive tumors generated in a mouse model of de novo carcinogenesis that closely recapitulates tumor initiation, progression and maintenance in vivo. Tumors were generated using the DMBA/TPA model of chemically induced carcinogenesis [1], tumor-bearing mice were subsequently treated with 5-FU, and tumor growth as well as response to treatment was monitored by measuring tumor volume twice a week. Based on these measurements, we selected two 5-FU resistant and two 5-FU sensitive tumors and performed whole transcriptome sequencing and in order to identify differentially expressed transcripts between the two sets. Data obtained is deposited and available through NCBI SRA (reference number SRP155180 - https://www.ncbi.nlm.nih.gov/sra/?term=SRP155180).

16.
Viruses ; 9(8)2017 08 12.
Article in English | MEDLINE | ID: mdl-28805675

ABSTRACT

Stem cells and cellular plasticity are likely important components of tissue response to infection. There is emerging evidence that stem cells harbor receptors for common pathogen motifs and that they are receptive to local inflammatory signals in ways suggesting that they are critical responders that determine the balance between health and disease. In the field of papillomaviruses stem cells have been speculated to play roles during the viral life cycle, particularly during maintenance, and virus-promoted carcinogenesis but little has been conclusively determined. I summarize here evidence that gives clues to the potential role of stem cells and cellular plasticity in the lifecycle papillomavirus and linked carcinogenesis. I also discuss outstanding questions which need to be resolved.


Subject(s)
Cell Plasticity , Papillomaviridae/physiology , Papillomavirus Infections/virology , Stem Cells/cytology , Animals , Humans , Papillomaviridae/genetics , Papillomavirus Infections/physiopathology , Stem Cells/virology , Virus Replication
17.
Front Cell Dev Biol ; 4: 118, 2016.
Article in English | MEDLINE | ID: mdl-27853732

ABSTRACT

The outcome of an inflammatory incident can hang in the balance between restoring health and tissue integrity on the one hand, and promoting aberrant tissue homeostasis and adverse outcomes on the other. Both microbial-related and sterile inflammation is a complex response characterized by a range of innate immune cell types, which produce and respond to cytokine mediators and other inflammatory signals. In turn, cells native to the tissue in question can sense these mediators and respond by migrating, proliferating and regenerating the tissue. In this review we will discuss how the specific outcomes of inflammatory incidents are affected by the direct regulation of stem cells and cellular plasticity. While less well appreciated than the effects of inflammatory signals on immune cells and other differentiated cells, the effects are crucial in understanding inflammation and appropriately managing therapeutic interventions.

19.
Stem Cell Res Ther ; 7(1): 98, 2016 Jul 29.
Article in English | MEDLINE | ID: mdl-27473056

ABSTRACT

The insufficient ability of specialized cells such as neurons, cardiac myocytes, and epidermal cells to regenerate after tissue damage poses a great challenge to treat devastating injuries and ailments. Recent studies demonstrated that a diverse array of cell types can be directly derived from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), or somatic cells by combinations of specific factors. The use of iPSCs and direct somatic cell fate conversion, or transdifferentiation, holds great promise for regenerative medicine as these techniques may circumvent obstacles related to immunological rejection and ethical considerations. However, producing iPSC-derived keratinocytes requires a lengthy two-step process of initially generating iPSCs and subsequently differentiating into skin cells, thereby elevating the risk of cellular damage accumulation and tumor formation. In this study, we describe the reprogramming of mouse embryonic fibroblasts into functional keratinocytes via the transient expression of pluripotency factors coupled with directed differentiation. The isolation of an iPSC intermediate is dispensable when using this method. Cells derived with this approach, termed induced keratinocytes (iKCs), morphologically resemble primary keratinocytes. Furthermore they express keratinocyte-specific markers, downregulate mesenchymal markers as well as the pluripotency factors Oct4, Sox2, and Klf4, and they show important functional characteristics of primary keratinocytes. iKCs can be further differentiated by high calcium administration in vitro and are capable of regenerating a fully stratified epidermis in vivo. Efficient conversion of somatic cells into keratinocytes could have important implications for studying genetic skin diseases and designing regenerative therapies to ameliorate devastating skin conditions.


Subject(s)
Cellular Reprogramming , Fibroblasts/cytology , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/transplantation , Regeneration/genetics , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation , Embryo, Mammalian , Epidermal Cells , Epidermis/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Nude , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transgenes , Tretinoin/pharmacology
20.
PLoS One ; 10(12): e0143840, 2015.
Article in English | MEDLINE | ID: mdl-26649886

ABSTRACT

Tumorigenesis is a complex, multistep process that depends on numerous alterations within the cell and contribution from the surrounding stroma. The ability to model macroscopic tumor evolution with high fidelity may contribute to better predictive tools for designing tumor therapy in the clinic. However, attempts to model tumor growth have mainly been developed and validated using data from xenograft mouse models, which fail to capture important aspects of tumorigenesis including tumor-initiating events and interactions with the immune system. In the present study, we investigate tumor growth and therapy dynamics in a mouse model of de novo carcinogenesis that closely recapitulates tumor initiation, progression and maintenance in vivo. We show that the rate of tumor growth and the effects of therapy are highly variable and mouse specific using a Gompertz model to describe tumor growth and a two-compartment pharmacokinetic/ pharmacodynamic model to describe the effects of therapy in mice treated with 5-FU. We show that inter-mouse growth variability is considerably larger than intra-mouse variability and that there is a correlation between tumor growth and drug kill rates. Our results show that in vivo tumor growth and regression in a double transgenic mouse model are highly variable both within and between subjects and that mathematical models can be used to capture the overall characteristics of this variability. In order for these models to become useful tools in the design of optimal therapy strategies and ultimately in clinical practice, a subject-specific modelling strategy is necessary, rather than approaches that are based on the average behavior of a given subject population which could provide erroneous results.


Subject(s)
Carcinogenesis , Cell Transformation, Neoplastic , Disease Models, Animal , Mice, Transgenic , Animals , Carcinogens , Humans , Mice , Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL