Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Nat Commun ; 15(1): 1038, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310103

ABSTRACT

There are significant commonalities among several pathologies involving fibroblasts, ranging from auto-immune diseases to fibrosis and cancer. Early steps in cancer development and progression are closely linked to fibroblast senescence and transformation into tumor-promoting cancer-associated fibroblasts (CAFs), suppressed by the androgen receptor (AR). Here, we identify ANKRD1 as a mesenchymal-specific transcriptional coregulator under direct AR negative control in human dermal fibroblasts (HDFs) and a key driver of CAF conversion, independent of cellular senescence. ANKRD1 expression in CAFs is associated with poor survival in HNSCC, lung, and cervical SCC patients, and controls a specific gene expression program of myofibroblast CAFs (my-CAFs). ANKRD1 binds to the regulatory region of my-CAF effector genes in concert with AP-1 transcription factors, and promotes c-JUN and FOS association. Targeting ANKRD1 disrupts AP-1 complex formation, reverses CAF activation, and blocks the pro-tumorigenic properties of CAFs in an orthotopic skin cancer model. ANKRD1 thus represents a target for fibroblast-directed therapy in cancer and potentially beyond.


Subject(s)
Cancer-Associated Fibroblasts , Skin Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Fibroblasts/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Skin Neoplasms/pathology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Tumor Microenvironment
2.
bioRxiv ; 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37425957

ABSTRACT

Alterations of nuclear structure and function, and associated impact on gene transcription, are a hallmark of cancer cells. Little is known of these alterations in Cancer-Associated Fibroblasts (CAFs), a key component of the tumor stroma. Here we show that loss of androgen receptor (AR), which triggers early steps of CAF activation in human dermal fibroblasts (HDFs), leads to nuclear membrane alterations and increased micronuclei formation, which are unlinked from induction of cellular senescence. Similar alterations occur in fully established CAFs, which are overcome by restored AR function. AR associates with nuclear lamin A/C and loss of AR results in a substantially increased lamin A/C nucleoplasmic redistribution. Mechanistically, AR functions as a bridge between lamin A/C with the protein phosphatase PPP1. In parallel with a decreased lamin-PPP1 association, AR loss results in a marked increase of lamin A/C phosphorylation at Ser 301, which is also a feature of CAFs. Phosphorylated lamin A/C at Ser 301 binds to the transcription promoter regulatory region of several CAF effector genes, which are upregulated due to the loss of AR. More directly, expression of a lamin A/C Ser301 phosphomimetic mutant alone is sufficient to convert normal fibroblasts into tumor-promoting CAFs of the myofibroblast subtype, without an impact on senescence. These findings highlight the pivotal role of the AR-lamin A/C-PPP1 axis and lamin A/C phosphorylation at Ser 301 in driving CAF activation.

3.
Science ; 376(6590): eabh1623, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35420948

ABSTRACT

Human cells produce thousands of lipids that change during cell differentiation and can vary across individual cells of the same type. However, we are only starting to characterize the function of these cell-to-cell differences in lipid composition. Here, we measured the lipidomes and transcriptomes of individual human dermal fibroblasts by coupling high-resolution mass spectrometry imaging with single-cell transcriptomics. We found that the cell-to-cell variations of specific lipid metabolic pathways contribute to the establishment of cell states involved in the organization of skin architecture. Sphingolipid composition is shown to define fibroblast subpopulations, with sphingolipid metabolic rewiring driving cell-state transitions. Therefore, cell-to-cell lipid heterogeneity affects the determination of cell states, adding a new regulatory component to the self-organization of multicellular systems.


Subject(s)
Fibroblasts , Skin , Sphingolipids , Fibroblasts/chemistry , Fibroblasts/classification , Fibroblasts/metabolism , Humans , Lipidomics/methods , Metabolic Networks and Pathways , Skin/chemistry , Skin/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Sphingolipids/analysis , Sphingolipids/metabolism , Transcriptome
4.
EMBO Rep ; 22(3): e52152, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33554445

ABSTRACT

"Flash forward genetics" refers to a genetic approach based on the functional interaction of a given factor with unknown partner(s) converging on shared targets across evolutionary boundaries. A study by Li et al (2021), published in this issue of EMBO Reports, illustrates the innovative potential of the approach. The authors applied it to identify interacting factors for FOXN1, a mammalian transcription factor with a highly specialized function in hair follicle morphogenesis and thymus. The authors express FOXN1 in the Drosophila eye to perform an unbiased genetic screen in a totally heterologous system. In a remarkable tour de force, the authors identify and characterize a factor so far known for its ubiquitous function in transcription elongation, AFF4. Li et al show that AFF4 plays also a specific role in hair follicle and thymus development in the mouse overlapping with that of FOXN1.


Subject(s)
Forkhead Transcription Factors , Hair Follicle , Animals , Biological Evolution , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Hair Follicle/metabolism , Mice , Mice, Nude
5.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33112375

ABSTRACT

Melanoma susceptibility differs significantly in male versus female populations. Low levels of androgen receptor (AR) in melanocytes of the two sexes are accompanied by heterogeneous expression at various stages of the disease. Irrespective of expression levels, genetic and pharmacological suppression of AR activity in melanoma cells blunts proliferation and induces senescence, while increased AR expression or activation exert opposite effects. AR down-modulation elicits a shared gene expression signature associated with better patient survival, related to interferon and cytokine signaling and DNA damage/repair. AR loss leads to dsDNA breakage, cytoplasmic leakage, and STING activation, with AR anchoring the DNA repair proteins Ku70/Ku80 to RNA Pol II and preventing RNA Pol II-associated DNA damage. AR down-modulation or pharmacological inhibition suppresses melanomagenesis, with increased intratumoral infiltration of macrophages and, in an immune-competent mouse model, cytotoxic T cells. AR provides an attractive target for improved management of melanoma independent of patient sex.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Melanoma/genetics , Receptors, Androgen/genetics , Signal Transduction/genetics , Animals , Carcinogenesis/pathology , Cell Line, Tumor , DNA Damage/genetics , DNA Repair/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , RNA Polymerase II/genetics
6.
Nat Commun ; 11(1): 5126, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046701

ABSTRACT

Cancer associated fibroblasts (CAFs) are a key component of the tumor microenvironment. Genomic alterations in these cells remain a point of contention. We report that CAFs from skin squamous cell carcinomas (SCCs) display chromosomal alterations, with heterogeneous NOTCH1 gene amplification and overexpression that also occur, to a lesser extent, in dermal fibroblasts of apparently unaffected skin. The fraction of the latter cells harboring NOTCH1 amplification is expanded by chronic UVA exposure, to which CAFs are resistant. The advantage conferred by NOTCH1 amplification and overexpression can be explained by NOTCH1 ability to block the DNA damage response (DDR) and ensuing growth arrest through suppression of ATM-FOXO3a association and downstream signaling cascade. In an orthotopic model of skin SCC, genetic or pharmacological inhibition of NOTCH1 activity suppresses cancer/stromal cells expansion. Here we show that NOTCH1 gene amplification and increased expression in CAFs are an attractive target for stroma-focused anti-cancer intervention.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Gene Amplification , Receptor, Notch1/metabolism , Skin Neoplasms/metabolism , Animals , DNA Damage , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Humans , Mice , Mice, SCID , Receptor, Notch1/genetics , Skin/metabolism , Skin Neoplasms/genetics
7.
EMBO Rep ; 21(2): e49924, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31930672

ABSTRACT

In times of fake news, post-truths and post-science, the principles of science can inform all kinds of inquiries into the true nature of reality.

8.
Cell Rep ; 28(9): 2358-2372.e6, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31461652

ABSTRACT

Heterogeneity of cancer-associated fibroblasts (CAFs) can result from activation of distinct signaling pathways. We show that in primary human dermal fibroblasts (HDFs), fibroblast growth factor (FGF) and transforming growth factor ß (TGF-ß) signaling oppositely modulate multiple CAF effector genes. Genetic abrogation or pharmacological inhibition of either pathway results in induction of genes responsive to the other, with the ETV1 transcription factor mediating the FGF effects. Duality of FGF/TGF-ß signaling and differential ETV1 expression occur in multiple CAF strains and fibroblasts of desmoplastic versus non-desmoplastic skin squamous cell carcinomas (SCCs). Functionally, HDFs with opposite TGF-ß versus FGF modulation converge on promoting cancer cell proliferation. However, HDFs with increased TGF-ß signaling enhance invasive properties and epithelial-mesenchymal transition (EMT) of SCC cells, whereas HDFs with increased FGF signaling promote macrophage infiltration. The findings point to a duality of FGF versus TGF-ß signaling in distinct CAF populations that promote cancer development through modulation of different processes.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/metabolism , DNA-Binding Proteins/metabolism , Fibroblast Growth Factors/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Child, Preschool , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Skin Neoplasms/pathology
10.
Autophagy ; 15(4): 738-739, 2019 04.
Article in English | MEDLINE | ID: mdl-30653445

ABSTRACT

In the tumor stroma, cancer-associated fibroblasts (CAFs) affect all aspects of tumor evolution. Whereas several programs leading to CAF activation have been elucidated, little is known about the impact of the microenvironment on the turnover of key CAF regulators. RBPJ/CSL is a transcriptional repressor that mediates NOTCH signaling and its down-modulation activates the gene expression program(s) leading to stromal senescence and CAF activation. We overview our evidence that conditions increasing macroautophagy/autophagy, as often found in the stroma of tumors, cause the down-modulation of the RBPJ protein. This event requires the autophagic machinery and is functionally relevant because it is associated with an increase of CAF effector gene expression. The mechanism involves the direct association with the autophagy receptor SQSTM1/p62, which is required for RBPJ down-modulation. As a reflection of increased autophagy in the stroma, both the RBPJ and SQSTM1 proteins are down-modulated in Squamous Cell Carcinoma (SCC) patient-derived CAFs. Increasing RBPJ cellular levels stabilizes SQSTM1 and down-modulates the autophagic process. Our findings identify an autophagy-initiated mechanism for RBPJ down-modulation leading to increased CAF gene expression.


Subject(s)
Autophagy , Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Fibroblasts , Humans , Signal Transduction , Tumor Microenvironment
11.
J Clin Invest ; 128(12): 5531-5548, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30395538

ABSTRACT

The aging-associated increase of cancer risk is linked with stromal fibroblast senescence and concomitant cancer-associated fibroblast (CAF) activation. Surprisingly little is known about the role of androgen receptor (AR) signaling in this context. We have found downmodulated AR expression in dermal fibroblasts underlying premalignant skin cancer lesions (actinic keratoses and dysplastic nevi) as well as in CAFs from the 3 major skin cancer types, squamous cell carcinomas (SCCs), basal cell carcinomas, and melanomas. Functionally, decreased AR expression in primary human dermal fibroblasts (HDFs) from multiple individuals induced early steps of CAF activation, and in an orthotopic skin cancer model, AR loss in HDFs enhanced tumorigenicity of SCC and melanoma cells. Forming a complex, AR converged with CSL/RBP-Jκ in transcriptional repression of key CAF effector genes. AR and CSL were positive determinants of each other's expression, with BET inhibitors, which counteract the effects of decreased CSL, restoring AR expression and activity in CAFs. Increased AR expression in these cells overcame the consequences of CSL loss and was by itself sufficient to block the growth and tumor-enhancing effects of CAFs on neighboring cancer cells. As such, the findings establish AR as a target for stroma-focused cancer chemoprevention and treatment.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Neoplasm Proteins/metabolism , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Skin Neoplasms/metabolism , Transcriptional Activation , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Neoplasm Proteins/genetics , Receptors, Androgen/genetics , Repressor Proteins/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
12.
Cell Rep ; 24(12): 3108-3114.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231994

ABSTRACT

Cancer-associated fibroblasts (CAFs) are important at all tumor stages. CSL/RBPJκ suppresses the gene expression program leading to CAF activation and associated metabolic reprogramming, as well as autophagy. Little is known about CSL protein turnover, especially in the tumor microenvironment. We report that, in human dermal fibroblasts (HDFs), conditions inducing autophagy-often found in tumor stroma-down-regulate CSL protein levels but do not affect its mRNA levels. Genetic or pharmacologic targeting of the autophagic machinery blocks CSL down-modulation. Mechanistically, endogenous CSL associates with the autophagy and signaling adaptor p62/SQSTM1, which is required for CSL down-modulation by autophagy. This is functionally significant, because both CSL and p62 levels are lower in skin cancer-derived CAFs, in which autophagy is increased. Increasing cellular CSL levels stabilizes p62 and down-modulates the autophagic process. We reveal here an autophagy-initiated mechanism for CSL down-modulation, which could be targeted for stroma-focused cancer prevention and treatment.


Subject(s)
Autophagy , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Sequestosome-1 Protein/metabolism , Skin Neoplasms/metabolism , Animals , Cell Line , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Protein Binding , Protein Stability , Sequestosome-1 Protein/genetics , Signal Transduction
13.
J Clin Invest ; 128(6): 2581-2599, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29757189

ABSTRACT

Notch 1/2 genes play tumor-suppressing functions in squamous cell carcinoma (SCC), a very common malignancy in skin and internal organs. In contrast with Notch, we show that the transcription factor CSL (also known as RBP-Jκ), a key effector of canonical Notch signaling endowed with intrinsic transcription-repressive functions, plays a tumor-promoting function in SCC development. Expression of this gene decreased in upper epidermal layers and human keratinocytes (HKCs) undergoing differentiation, while it increased in premalignant and malignant SCC lesions from skin, head/neck, and lung. Increased CSL levels enhanced the proliferative potential of HKCs and SCC cells, while silencing of CSL induced growth arrest and apoptosis. In vivo, SCC cells with increased CSL levels gave rise to rapidly expanding tumors, while cells with silenced CSL formed smaller and more differentiated tumors with enhanced inflammatory infiltrate. Global transcriptomic analysis of HKCs and SCC cells with silenced CSL revealed major modulation of apoptotic, cell-cycle, and proinflammatory genes. We also show that the histone demethylase KDM6B is a direct CSL-negative target, with inverse roles of CSL in HKC and SCC proliferative capacity, tumorigenesis, and tumor-associated inflammatory reaction. CSL/KDM6B protein expression could be used as a biomarker of SCC development and indicator of cancer treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Signal Transduction , Animals , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism
14.
Cell Rep ; 20(10): 2468-2479, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877478

ABSTRACT

The connection between signaling pathways activating cancer-associated fibroblasts (CAFs) remains to be determined. Metabolic alterations linked to autophagy have also been implicated in CAF activation. CSL/RBPJ, a transcriptional repressor that mediates Notch signaling, suppresses the gene expression program(s), leading to stromal senescence and CAF activation. Deregulated GLI signaling can also contribute to CAF conversion. Here, we report that compromised CSL function depends on GLI activation for conversion of human dermal fibroblasts into CAFs, separately from cellular senescence. Decreased CSL upregulates the expression of the ULK3 kinase, which binds and activates GLI2. Increased ULK3 also induces autophagy, which is unlinked from GLI and CAF activation. ULK3 upregulation occurs in the CAFs of several tumor types, and ULK3 silencing suppresses the tumor-enhancing properties of these cells. Thus, ULK3 links two key signaling pathways involved in CAF conversion and is an attractive target for stroma-focused anti-cancer intervention.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Fibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Zinc Finger Protein GLI1/metabolism , Animals , Autophagy/physiology , Female , Fluorescent Antibody Technique , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Zinc Finger Protein GLI1/genetics
15.
J Exp Med ; 214(8): 2349-2368, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28684431

ABSTRACT

Cancer-associated fibroblasts (CAFs) are important for tumor initiation and promotion. CSL, a transcriptional repressor and Notch mediator, suppresses CAF activation. Like CSL, ATF3, a stress-responsive transcriptional repressor, is down-modulated in skin cancer stromal cells, and Atf3 knockout mice develop aggressive chemically induced skin tumors with enhanced CAF activation. Even at low basal levels, ATF3 converges with CSL in global chromatin control, binding to few genomic sites at a large distance from target genes. Consistent with this mode of regulation, deletion of one such site 2 Mb upstream of IL6 induces expression of the gene. Observed changes are of translational significance, as bromodomain and extra-terminal (BET) inhibitors, unlinking activated chromatin from basic transcription, counteract the effects of ATF3 or CSL loss on global gene expression and suppress CAF tumor-promoting properties in an in vivo model of squamous cancer-stromal cell expansion. Thus, ATF3 converges with CSL in negative control of CAF activation with epigenetic changes amenable to cancer- and stroma-focused intervention.


Subject(s)
Activating Transcription Factor 3/physiology , Cancer-Associated Fibroblasts/physiology , Chromatin/physiology , Muscle Proteins/physiology , Animals , Carcinoma, Squamous Cell/physiopathology , Keratinocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/physiopathology
16.
Oncotarget ; 7(37): 58717-58727, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27542230

ABSTRACT

The Notch/CSL pathway plays an important role in skin homeostasis and carcinogenesis. CSL, the key effector of canonical Notch signaling endowed with an intrinsic transcription repressive function, suppresses stromal fibroblast senescence and Cancer Associated Fibroblast (CAF) activation through direct down-modulation of key effector genes. Interacting proteins that participate with CSL in this context are as yet to be identified. We report here that Programmed Cell Death 4 (PDCD4), a nuclear/cytoplasmic shuttling protein with multiple functions, associates with CSL and plays a similar role in suppressing dermal fibroblast senescence and CAF activation. Like CSL, PDCD4 is down-regulated in stromal fibroblasts of premalignant skin actinic keratosis (AKs) lesions and squamous cell carcinoma (SCC). While devoid of intrinsic DNA binding capability, PDCD4 is present at CSL binding sites of CAF marker genes as well as canonical Notch/CSL targets and suppresses expression of these genes in a fibroblast-specific manner. Thus, we propose that PDCD4 is part of the CSL repressive complex involved in negative control of stromal fibroblasts conversion into CAFs.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cancer-Associated Fibroblasts/immunology , Carcinoma, Squamous Cell/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Keratosis, Actinic/metabolism , RNA-Binding Proteins/metabolism , Skin Neoplasms/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cellular Senescence , Down-Regulation , HEK293 Cells , HeLa Cells , Humans , Keratosis, Actinic/genetics , Mice , Mice, SCID , Protein Binding , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Skin Neoplasms/genetics , Transcription, Genetic , Xenograft Model Antitumor Assays
18.
Oncotarget ; 7(30): 48011-48026, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27384993

ABSTRACT

Adenocarcinomas of the prostate arise as multifocal heterogeneous lesions as the likely result of genetic and epigenetic alterations and deranged cell-cell communication. Notch signaling is an important form of intercellular communication with a role in growth/differentiation control and tumorigenesis. Contrasting reports exist in the literature on the role of this pathway in prostate cancer (PCa) development. We show here that i) compared to normal prostate tissue, Notch1 expression is significantly reduced in a substantial fraction of human PCas while it is unaffected or even increased in others; ii) acute Notch activation both inhibits and induces process networks associated with prostatic neoplasms; iii) down-modulation of Notch1 expression and activity in immortalized normal prostate epithelial cells increases their proliferation potential, while increased Notch1 activity in PCa cells suppresses growth and tumorigenicity through a Smad3-dependent mechanism involving p21WAF1/CIP1; iv) prostate cancer cells resistant to Notch growth inhibitory effects retain Notch1-induced upregulation of pro-oncogenic genes, like EPAS1 and CXCL6, also overexpressed in human PCas with high Notch1 levels. Taken together, these results reconcile conflicting data on the role of Notch1 in prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Aged , Carcinogenesis , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Genes, Tumor Suppressor , Humans , Male , Middle Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, Notch1/genetics , Signal Transduction
19.
Cell Cycle ; 15(13): 1767-78, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27163456

ABSTRACT

CSL is a key transcriptional repressor and mediator of Notch signaling. Despite wide interest in CSL, mechanisms responsible for its own regulation are little studied. CSL down-modulation in human dermal fibroblasts (HDFs) leads to conversion into cancer associated fibroblasts (CAF), promoting keratinocyte tumors. We show here that CSL transcript levels differ among HDF strains from different individuals, with negative correlation with genes involved in DNA damage/repair. CSL expression is negatively regulated by stress/DNA damage caused by UVA, Reactive Oxygen Species (ROS), smoke extract, and doxorubicin treatment. P53, a key effector of the DNA damage response, negatively controls CSL gene transcription, through suppression of CSL promoter activity and, indirectly, by increased p21 expression. CSL was previously shown to bind p53 suppressing its activity. The present findings indicate that p53, in turn, decreases CSL expression, which can serve to enhance p53 activity in acute DNA damage response of cells.


Subject(s)
DNA Damage/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Oxidative Stress/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Cell Line , Dermis/cytology , Fibroblasts/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Male , Mutagens/toxicity , Oxidative Stress/drug effects , Receptors, Notch/metabolism , Signal Transduction/genetics
20.
Cancer Cell ; 29(5): 622-637, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27165741

ABSTRACT

Squamous cell carcinomas (SCCs) represent the most frequent human solid tumors and are a major cause of cancer mortality. These highly heterogeneous tumors arise from closely interconnected epithelial cell populations with intrinsic self-renewal potential inversely related to the stratified differentiation program. SCCs can also originate from simple or pseudo-stratified epithelia through activation of quiescent cells and/or a switch in cell-fate determination. Here, we focus on specific determinants implicated in the development of SCCs by recent large-scale genomic, genetic, and epigenetic studies, and complementary functional analysis. The evidence indicates that SCCs from various body sites, while clinically treated as separate entities, have common determinants, pointing to a unified perspective of the disease and potential new avenues for prevention and treatment.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , Mutation , Alcohol Drinking/adverse effects , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/therapy , Humans , Infections/complications , Models, Genetic , Smoking/adverse effects , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...