Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 10(1): 99, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402225

ABSTRACT

Cancer most frequently develops in self-renewal tissues that are the target of genetic alterations due to mutagens or intrinsic DNA replication errors. Histone γH2AX has a critical role in the cellular DNA repair pathway cascade and contributes to genomic stability. However, the role of γH2AX in the ontology of cancer is unclear. We have investigated this issue in the epidermis, a self-renewal epithelium continuously exposed to genetic hazard and replication stress. Silencing H2AX caused cell cycle hyperactivation, impaired DNA repair and epidermal hyperplasia in the skin. However, mutagen-induced carcinogenesis was strikingly reduced in the absence of H2AX. KO tumours appeared significantly later than controls and were fewer, smaller and more benign. The stem cell marker Δp63 drastically diminished in the KO epidermis. We conclude that H2AX is required for tissue-making during both homoeostasis and tumourigenesis, possibly by contributing to the control and repair of stem cells. Therefore, although H2AX is thought to act as a tumour suppressor and our results show that it contributes to homeostasis, they also indicate that it is required for the development of cancer.

2.
Cell Death Discov ; 9(1): 21, 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36681661

ABSTRACT

Epithelial transdifferentiation is frequent in tissue hyperplasia and contributes to disease in various degrees. Squamous metaplasia (SQM) precedes epidermoid lung cancer, an aggressive and frequent malignancy, but it is rare in the epithelium of the mammary gland. The mechanisms leading to SQM in the lung have been very poorly investigated. We have studied this issue on human freshly isolated cells and organoids. Here we show that human lung or mammary cells strikingly undergo SQM with polyploidisation when they are exposed to genotoxic or mitotic drugs, such as Doxorubicin or the cigarette carcinogen DMBA, Nocodazole, Taxol or inhibitors of Aurora-B kinase or Polo-like kinase. To note, the epidermoid response was attenuated when DNA repair was enhanced by Enoxacin or when mitotic checkpoints where abrogated by inhibition of Chk1 and Chk2. The results show that DNA damage has the potential to drive SQM via mitotic checkpoints, thus providing novel molecular candidate targets to tackle lung SCC. Our findings might also explain why SCC is frequent in the lung, but not in the mammary gland and why chemotherapy often causes complicating skin toxicity.

3.
Int J Nanomedicine ; 17: 5747-5760, 2022.
Article in English | MEDLINE | ID: mdl-36466783

ABSTRACT

Introduction: A great challenge in nanomedicine, and more specifically in theranostics, is to improve the specificity, selectivity, and targeting of nanomaterials towards target tissues or cells. The topical use of nanomedicines as adjuvants to systemic chemotherapy can significantly improve the survival of patients affected by localized carcinomas, reducing the side effects of traditional drugs and preventing local recurrences. Methods: Here, we have used the Shiga toxin, to design a safe, high-affinity protein-ligand (ShTxB) to bind the globotriaosylceramide receptor (GB3) that is overexpressed on the surfaces of preneoplastic and malignant cancer cells in the head and neck tumors. Results: We find that ShTxB functionalized gold nanorods are efficiently retrotranslocated to the GB3-positive cell cytoplasms. After 3 minutes of laser radiation with a wavelength resonant with the AuNR longitudinal localized surface plasmon, the death of the targeted cancer cells is activated. Both preclinical murine models and patient biopsy cells show the non-cytotoxic nature of these functionalized nanoparticles before light activation and their treatment selectivity. Discussion: These results show how the use of nanomedicines directed by natural ligands can represent an effective treatment for aggressive localized cancers, such as squamous cell carcinoma of the oral cavity.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Nanotubes , Humans , Animals , Mice , Gold , Shiga Toxin , Mouth Neoplasms/drug therapy
4.
Orphanet J Rare Dis ; 17(1): 345, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36068614

ABSTRACT

BACKGROUND: Novel developmental mutations associated with disease are a continuous challenge in medicine. Clinical consequences caused by these mutations include neuron and cognitive alterations that can lead to epilepsy or autism spectrum disorders. Often, it is difficult to identify the physiological defects and the appropriate treatments. RESULTS: We have isolated and cultured primary cells from the skin of a patient with combined epilepsy and autism syndrome. A mutation in the potassium channel protein Kv10.2 was identified. We have characterised the alteration of the mutant channel and found that it causes loss of function (LOF). Primary cells from the skin displayed a very striking growth defect and increased differentiation. In vitro treatment with various carbonic anhydrase inhibitors with various degrees of specificity for potassium channels, (Brinzolamide, Acetazolamide, Retigabine) restored the activation capacity of the mutated channel. Interestingly, the drugs also recovered in vitro the expansion capacity of the mutated skin cells. Furthermore, treatment with Acetazolamide clearly improved the patient regarding epilepsy and cognitive skills. When the treatment was temporarily halted the syndrome worsened again. CONCLUSIONS: By in vitro studying primary cells from the patient and the activation capacity of the mutated protein, we could first, find a readout for the cellular defects and second, test pharmaceutical treatments that proved to be beneficial. The results show the involvement of a novel LOF mutation of a Potassium channel in autism syndrome with epilepsy and the great potential of in vitro cultures of primary cells in personalised medicine of rare diseases.


Subject(s)
Autistic Disorder , Epilepsy , Potassium Channels, Voltage-Gated , Acetazolamide , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/metabolism , Humans , Mutation/genetics , Potassium Channels/genetics , Potassium Channels, Voltage-Gated/genetics
6.
Cancer Gene Ther ; 29(8-9): 1160-1167, 2022 08.
Article in English | MEDLINE | ID: mdl-35022520

ABSTRACT

In spite of extensive research and advances on the molecular biology of melanoma, the process of melanocytic differentiation or its relationship with proliferation is poorly understood. The role of proto-oncogenes in normal melanocyte biology is also intriguing. Proto-oncogene MYC is overexpressed in 40% of melanomas. It has been suggested that MYC can mediate senescence bypass in malignant melanocytes, an important event in melanoma development, likely in cooperation with other oncogenic pathways. However, despite the apparent importance of MYC in melanoma, its functions in normal melanocytes are unknown. We have overexpressed MYC in freshly isolated human primary melanocytes and studied the effects on melanocytic proliferation and differentiation. MYC promoted a transient activation of melanocytes including cell cycle entry, DNA damage and cell migration. Subsequently, MYC induced melanogenesis, increased cellular size and complexity and senescence. Interestingly, we also found strong expression of MYC in regions of human nevi displaying high pigmentation and high expression of senescence marker p16. The results altogether show that MYC drives melanocytic differentiation and suggest that senescence is associated with differentiation. We discuss the implications into the mechanisms governing melanocytic differentiation and the development of melanoma.


Subject(s)
Melanoma , Skin Neoplasms , Cell Differentiation/genetics , Cellular Senescence/genetics , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Proto-Oncogenes , Skin Neoplasms/genetics
7.
Sci Rep ; 11(1): 24426, 2021 12 24.
Article in English | MEDLINE | ID: mdl-34952907

ABSTRACT

Allergies to grass pollen affects about 20% of the population worldwide. In the last few decades, the South American grass Cortaderia selloana (CS, Pampas grass) has expanded worldwide in a variety of countries including the USA, Australia and Western Europe. In many of these locations, CS has strikingly spread and has now been classified an invasive species. Many pernicious consequences of CS have been reported for local biodiversity, landscape and structures. However, the effect on human health has not been studied. To investigate this issue, we have chosen a European region on the northern cost of Spain where CS spread is overwhelming, Cantabria. We obtained CS pollen extract and analysed the allergenic reaction of 98 patients that were allergic to pollen of local grasses. We determined the skin reaction and the presence of specific IgE antibodies (sIgE) to CS or to a typical autochthonous grass, Phleum pratense. We also compared the seasonal symptoms with reported grass pollen counts in the area. The results strongly suggest that CS can cause respiratory allergies at a similar extent to the local grasses. Given that CS pollinises later than the local grasses, this would extend the period of grass allergies in the region for about three months every year, as stated by most of the patients. This is the first study reported on the effects of the striking expansion of CS on human health. Considering the strong impact that respiratory allergies have on the population, our results suggest that CS can currently constitute a relevant environmental health issue.


Subject(s)
Allergens/immunology , Hypersensitivity , Poaceae/immunology , Pollen/immunology , Rhinitis, Allergic, Seasonal , Humans , Hypersensitivity/epidemiology , Hypersensitivity/immunology , Immunoglobulin E/blood , Rhinitis, Allergic, Seasonal/epidemiology , Rhinitis, Allergic, Seasonal/immunology , Spain/epidemiology
8.
Oncogene ; 40(1): 152-162, 2021 01.
Article in English | MEDLINE | ID: mdl-33097856

ABSTRACT

The control of cell fate is critical to homeostasis and cancer. Cell cycle cdk inhibitor p21CIP1 has a central and paradoxical role in the regulatory crossroads leading to senescence, apoptosis, or differentiation. p21 is an essential target of tumor suppressor p53, but it also is regulated independently. In squamous self-renewal epithelia continuously exposed to mutagenesis, p21 controls cell fate by mechanisms still intriguing. We previously identified a novel epidermoid DNA damage-differentiation response. We here show that p21 intervenes in the mitosis block that is required for the squamous differentiation response to cell cycle deregulation and replication stress. The inactivation of endogenous p21 in human primary keratinocytes alleviated the differentiation response to oncogenic loss of p53 or overexpression of the DNA replication major regulator Cyclin E. The bypass of p21-induced mitotic block involving upregulation of Cyclin B allowed DNA damaged cells to escape differentiation and continue to proliferate. In addition, loss of p21 drove keratinocytes from differentiation to apoptosis upon moderate UV irradiation. The results show that p21 is required to drive keratinocytes towards differentiation in response to genomic stress and shed light into its dual and paradoxical role in carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Keratinocytes/cytology , Animals , Apoptosis , Carcinoma, Squamous Cell/genetics , Cell Cycle , Cell Differentiation , Cells, Cultured , Cellular Senescence , Cyclin E/genetics , DNA Damage , DNA Replication , Gene Knockdown Techniques , HeLa Cells , Humans , Keratinocytes/metabolism , Mice , Primary Cell Culture , Tumor Suppressor Protein p53/genetics
9.
J Cell Biol ; 219(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-33007086

ABSTRACT

How rapid cell multiplication leads to cell differentiation in developing tissues is still enigmatic. This question is central to morphogenesis, cell number control, and homeostasis. Self-renewal epidermoid epithelia are continuously exposed to mutagens and are the most common target of cancer. Unknown mechanisms commit rapidly proliferating cells to post-mitotic terminal differentiation. We have over-activated or inhibited the endogenous DNA damage response (DDR) pathways by combinations of activating TopBP1 protein, specific shRNAs, or chemical inhibitors for ATR, ATM, and/or DNA-PK. The results dissect and demonstrate that these signals control keratinocyte differentiation in proliferating cells independently of actual DNA damage. The DDR limits keratinocyte multiplication upon hyperproliferative stimuli. Moreover, knocking down H2AX, a common target of the DDR pathways, inhibits the epidermoid phenotype. The results altogether show that the DDR is required to maintain the balance proliferation differentiation and suggest that is part of the squamous program. We propose a homeostatic model where genetic damage is automatically and continuously cleansed by cell-autonomous mechanisms.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Differentiation , Cell Proliferation , DNA Damage , Epithelial Cells/cytology , Keratinocytes/cytology , Epithelial Cells/metabolism , Histones , Humans , Keratinocytes/metabolism , Phosphorylation , Signal Transduction
10.
Cell Death Differ ; 27(8): 2451-2467, 2020 08.
Article in English | MEDLINE | ID: mdl-32080348

ABSTRACT

The cellular mechanisms controlling cell fate in self-renewal tissues remain unclear. Cell cycle failure often leads to an apoptosis anti-oncogenic response. We have inactivated Cdk1 or Polo-like-1 kinases, essential targets of the mitotic checkpoints, in the epithelia of skin and oral mucosa. Here, we show that inactivation of the mitotic kinases leading to polyploidy in vivo, produces a fully differentiated epithelium. Cells within the basal layer aberrantly differentiate and contain large or various nuclei. Freshly isolated KO cells were also differentiated and polyploid. However, sustained metaphase arrest downstream of the spindle anaphase checkpoint (SAC) due to abrogation of CDC20 (essential cofactor of anaphase-promoting complex), impaired squamous differentiation and resulted in apoptosis. Therefore, upon prolonged arrest keratinocytes need to slip beyond G2 or mitosis in order to initiate differentiation. The results altogether demonstrate that mitotic checkpoints drive squamous cell fate towards differentiation or apoptosis in response to genetic damage.


Subject(s)
Apoptosis , Cell Differentiation , Epithelium/pathology , G2 Phase , Mitosis , Animals , CDC2 Protein Kinase/metabolism , Cdc20 Proteins/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Cytokinesis , Epidermis/pathology , Humans , Hyperplasia , Mice , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
11.
Methods Mol Biol ; 2109: 83-92, 2020.
Article in English | MEDLINE | ID: mdl-31123997

ABSTRACT

The epidermis is continuously exposed to environmental hazard and undergoes continuous cell renewal. The maintenance of the epidermal balance between proliferation and differentiation is essential for the homeostasis of the skin. Proliferation and terminal differentiation are compartmentalized in basal and suprabasal layers, respectively. These compartments can be identified by different patterns of protein expression that can be used as differentiation markers. For instance, components of the intermediate filament cytoskeleton keratins K5 and K14 are confined to the proliferative basal layer, while keratins K1 and K10, keratins K6 and K16, or precursors of the cornified envelope such as involucrin are expressed by suprabasal terminally differentiating cells. The analysis of the expression of these markers allows studying the imbalance typical of disease. Although these markers have been traditionally analyzed on skin microsections, on attached cells by immunostaining or by western blotting, it is possible and advantageous to quantify them by flow cytometry. We have extensively applied this technology onto human and mouse keratinocytes. Here we describe detailed flow cytometry methods to determine the differentiation status of keratinocyte populations.


Subject(s)
Biomarkers/metabolism , Keratinocytes/cytology , Keratins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Flow Cytometry , Humans , Keratinocytes/metabolism
12.
Methods Mol Biol ; 2109: 113-123, 2020.
Article in English | MEDLINE | ID: mdl-31123998

ABSTRACT

Keratinocytes are hard to transfect. Viral vectors are a good alternative to genetically modify primary keratinocytes. A classical method is the use of retroviral vectors by co-culture of keratinocytes with virus-producer cells. This method is efficient in high-calcium conditions with feeder cells. However, sometimes co-culture is not possible and is more laborious as producer cells need to be replaced by feeder cells. Our solution is the use of lentiviral vectors, far more efficient as supernatant on keratinocytes. In this chapter we describe improved detailed protocols for stable genetic modification of human primary keratinocytes of the skin or head and neck, in both low- and high-calcium conditions by lentiviral vectors.


Subject(s)
Keratinocytes/cytology , Lentivirus/physiology , Primary Cell Culture/methods , 3T3 Cells , Animals , Calcium/metabolism , Coculture Techniques , Culture Media/chemistry , Feeder Cells/cytology , Humans , Keratinocytes/chemistry , Lentivirus/genetics , Mice , Transduction, Genetic
13.
Sci Rep ; 9(1): 736, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679581

ABSTRACT

Mutations in ADNP have been recently associated with intellectual disability and autism spectrum disorder. However, the clinical features of patients with this syndrome are not fully identified, and no treatment currently exists for these patients. Here, we extended the ADNP syndrome phenotype describing skin abnormalities in both a patient with ADNP syndrome and an Adnp haploinsufficient mice. The patient displayed thin dermis, hyperkeratotic lesions in periarticular areas and delayed wound healing. Patient-derived skin keratinocytes showed reduced proliferation and increased differentiation. Additionally, detection of cell cycle markers indicated that mutant cells exhibited impaired cell cycle progression. Treatment of ADNP-deficient keratinocytes with the ADNP-derived NAP peptide significantly reduced the expression of differentiation markers. Sonography and immunofluorescence staining of epidermal layers revealed that the dermis was thinner in the patient than in a healthy control. Adnp haploinsufficient mice (Adnp+/-) mimicked the human condition showing reduced dermal thickness. Intranasal administration of NAP significantly increased dermal thickness and normalized the levels of cell cycle and differentiation markers. Our observations provide a novel activity of the autism-linked ADNP in the skin that may serve to define the clinical phenotype of patients with ADNP syndrome and provide an attractive therapeutic option for skin alterations in these patients.


Subject(s)
Autism Spectrum Disorder/genetics , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Skin Abnormalities/genetics , Animals , Autism Spectrum Disorder/pathology , Cell Cycle/genetics , Cell Differentiation/genetics , Disease Models, Animal , Haploinsufficiency/genetics , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Keratinocytes/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Skin Abnormalities/pathology , Wound Healing/genetics
15.
Head Neck ; 40(11): 2487-2497, 2018 11.
Article in English | MEDLINE | ID: mdl-30311985

ABSTRACT

BACKGROUND: Squamous epithelia of the head and neck undergo continuous cell renewal and are continuously exposed to mutagenic hazard, the main cause of cancer. How they maintain homeostasis upon cell cycle deregulation is unclear. METHODS: To elucidate how head and neck epithelia respond to cell cycle stress, we studied human keratinocytes from various locations (oral mucosa, tonsil, pharynx, larynx, and trachea). We made use of genotoxic or mitotic drugs (doxorubicin [DOXO], paclitaxel, and nocodazole), or chemical inhibitors of the mitotic checkpoint kinases, Aurora B and polo-like-1. We further tested the response to inactivation of p53, ectopic cyclin E, or to the chemical carcinogen 7,12-dimethylbenz[a]anthracene (DMBA). RESULTS: All treatments provoked DNA damage or mitosis impairment and strikingly triggered squamous differentiation and polyploidization, resulting in irreversible loss of clonogenic capacity. CONCLUSION: Keratinocytes from head and neck epithelia share a cell-autonomous squamous DNA damage-differentiation response that is common to the epidermis and might continuously protect them from cancer.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Proliferation/genetics , DNA Damage/drug effects , DNA Damage/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Biopsy, Needle , Cell Proliferation/drug effects , Cells, Cultured , Cyclin E/genetics , Doxorubicin/pharmacology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Male , Nocodazole/pharmacology , Oncogene Proteins/genetics , Paclitaxel/pharmacology , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity
16.
Cell Death Dis ; 9(11): 1094, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361544

ABSTRACT

The epidermis is a self-renewal epithelium continuously exposed to the genotoxic effects of ultraviolet (UV) light, the main cause of skin cancer. Therefore, it needs robust self-protective mechanisms facing genomic damage. p53 has been shown to mediate apoptosis in sunburn cells of the epidermis. However, epidermal cells daily receive sublethal mutagenic doses of UV and massive apoptosis would be deleterious. We have recently unravelled an anti-oncogenic keratinocyte DNA damage-differentiation response to cell cycle stress. We now have studied this response to high or moderate single doses of UV irradiation. Whereas, as expected, high levels of UV induced p53-dependent apoptosis, moderate levels triggered squamous differentiation. UV-induced differentiation was not mediated by endogenous p53. Overexpression of the mitosis global regulator FOXM1 alleviated the proliferative loss caused by UV. Conversely, knocking-down the mitotic checkpoint protein Wee1 drove UV-induced differentiation into apoptosis. Therefore, the results indicate that mitosis checkpoints determine the response to UV irradiation. The differentiation response was also found in cells of head and neck epithelia thus uncovering a common regulation in squamous tissues upon chronic exposure to mutagens, with implications into homeostasis and disease.


Subject(s)
Cell Differentiation/radiation effects , DNA Damage/radiation effects , Keratinocytes/metabolism , Mitosis/radiation effects , Radiation Dosage , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays/adverse effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/etiology , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins/genetics , Cells, Cultured , Gene Knockdown Techniques , HEK293 Cells , Humans , Keratin-13/metabolism , Protein-Tyrosine Kinases/genetics , Skin Neoplasms/etiology , Transfection
17.
Cancers (Basel) ; 10(9)2018 Sep 17.
Article in English | MEDLINE | ID: mdl-30227608

ABSTRACT

This study investigates for the first time the crosstalk between stromal fibroblasts and cancer stem cell (CSC) biology in head and neck squamous cell carcinomas (HNSCC), with the ultimate goal of identifying effective therapeutic targets. The effects of conditioned media from cancer-associated fibroblasts (CAFs) and normal fibroblasts (NFs) on the CSC phenotype were assessed by combining functional and expression analyses in HNSCC-derived cell lines. Further characterization of CAFs and NFs secretomes by mass spectrometry was followed by pharmacologic target inhibition. We demonstrate that factors secreted by CAFs but not NFs, in the absence of serum/supplements, robustly increased anchorage-independent growth, tumorsphere formation, and CSC-marker expression. Modulators of epidermal growth factor receptor (EGFR), insulin-like growth factor receptor (IGFR), and platelet-derived growth factor receptor (PDGFR) activity were identified as paracrine cytokines/factors differentially secreted between CAFs and NFs, in a mass spectrometry analysis. Furthermore, pharmacologic inhibition of EGFR, IGFR, and PDGFR significantly reduced CAF-induced tumorsphere formation and anchorage-independent growth suggesting a role of these receptor tyrosine kinases in sustaining the CSC phenotype. These findings provide novel insights into tumor stroma⁻CSC communication, and potential therapeutic targets to effectively block the CAF-enhanced CSC niche signaling circuit.

18.
Cell Death Differ ; 25(3): 471-476, 2018 03.
Article in English | MEDLINE | ID: mdl-29352263

ABSTRACT

Among the most intriguing and relevant questions in physiology is how developing tissues correctly coordinate proliferation with differentiation. Endoreplication, in a broad sense, is a consequence of a cell division block in the presence of an active cell cycle, and it typically occurs as cells differentiate terminally to fulfill a specialised function. Until recently, endoreplication was thought to be a rare variation of the cell cycle in mammals, more common in invertebrates and plants. However, in the last years, endoreplication has been uncovered in various tissues in mammalian organisms, including human. A recent report showing that cells in the mammary gland become binucleate at lactation sheds new insight into the importance of mammalian polyploidisation. We here propose that endoreplication is a widespread phenomenon in mammalian developing tissues that results from an automatic, robust and simple self-limiting mechanism coordinating cell multiplication with differentiation. This mechanism might act as a developmental timer. The model has implications for homeostasis control and carcinogenesis.


Subject(s)
Endoreduplication , Animals , Carcinogenesis , Cell Differentiation , Cell Proliferation , Homeostasis , Humans , Time Factors
19.
Cell Death Dis ; 8(6): e2901, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28661481

ABSTRACT

Squamous cell carcinoma (SCC) or epidermoid cancer is a frequent and aggressive malignancy. However in apparent paradox it retains the squamous differentiation phenotype except for very dysplastic lesions. We have shown that cell cycle stress in normal epidermal keratinocytes triggers a squamous differentiation response involving irreversible mitosis block and polyploidisation. Here we show that cutaneous SCC cells conserve a partial squamous DNA damage-induced differentiation response that allows them to overcome the cell division block. The capacity to divide in spite of drug-induced mitotic stress and DNA damage made well-differentiated SCC cells more genomically instable and more malignant in vivo. Consistently, in a series of human biopsies, non-metastatic SCCs displayed a higher degree of chromosomal alterations and higher expression of the S phase regulator Cyclin E and the DNA damage signal γH2AX than the less aggressive, non-squamous, basal cell carcinomas. However, metastatic SCCs lost the γH2AX signal and Cyclin E, or accumulated cytoplasmic Cyclin E. Conversely, inhibition of endogenous Cyclin E in well-differentiated SCC cells interfered with the squamous phenotype. The results suggest a dual role of cell cycle stress-induced differentiation in squamous cancer: the resulting mitotic blocks would impose, when irreversible, a proliferative barrier, when reversible, a source of genomic instability, thus contributing to malignancy.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Cyclin E/genetics , Histones/genetics , Skin Neoplasms/genetics , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , DNA Damage/drug effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Genomic Instability/genetics , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Mitosis/drug effects , Mitosis/genetics , Polyploidy , Primary Cell Culture , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology
20.
Eur J Cell Biol ; 96(6): 632-641, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28413121

ABSTRACT

Human hair follicle (HF) growth and hair shaft formation require terminal differentiation-associated cell cycle arrest of highly proliferative matrix keratinocytes. However, the regulation of this complex event remains unknown. CIP/KIP family member proteins (p21CIP1, p27KIP1 and p57KIP2) regulate cell cycle progression/arrest, endoreplication, differentiation and apoptosis. Since they have not yet been adequately characterized in the human HF, we asked whether and where CIP/KIP proteins localise in the human hair matrix and pre-cortex in relation to cell cycle activity and HF-specific epithelial cell differentiation that is marked by keratin 85 (K85) protein expression. K85 expression coincided with loss or reduction in cell cycle activity markers, including in situ DNA synthesis (EdU incorporation), Ki-67, phospho-histone H3 and cyclins A and B1, affirming a post-mitotic state of pre-cortical HF keratinocytes. Expression of CIP/KIP proteins was found abundantly within the proliferative hair matrix, concomitant with a role in cell cycle checkpoint control. p21CIP1, p27KIP1 and cyclin E persisted within post-mitotic keratinocytes of the pre-cortex, whereas p57KIP2 protein decreased but became nuclear. These data imply a supportive role for CIP/KIP proteins in maintaining proliferative arrest, differentiation and anti-apoptotic pathways, promoting continuous hair bulb growth and hair shaft formation in anagen VI. Moreover, post-mitotic hair matrix regions contained cells with enlarged nuclei, and DNA in situ hybridisation showed cells that were >2N in the pre-cortex. This suggests that CIP/KIP proteins might counterbalance cyclin E to control further rounds of DNA replication in a cell population that has a propensity to become tetraploid. These data shed new light on the in situ-biography of human hair matrix keratinocytes on their path of active cell cycling, arrest and terminal differentiation, and showcase the human HF as an excellent, clinically relevant model system for cell cycle physiology research of human epithelial cells within their natural tissue habitat.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Hair Follicle/growth & development , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , Hair Follicle/metabolism , Humans , Keratinocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...