Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
J Biophotonics ; 8(10): 795-803, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25411997

ABSTRACT

Low-level laser therapy (LLLT) has been extensively employed to improve epithelial wound healing, though the exact response of epithelium maturation and stratification after LLLT is unknown. Thus, this study aimed to assess the in vitro growth and differentiation of keratinocytes (KCs) and in vivo wound healing response when treated with LLLT. Human KCs (HaCaT cells) showed an enhanced proliferation with all the employed laser energy densities (3, 6 and 12 J/cm(2) , 660 nm, 100 mW), together with an increased expression of Cyclin D1. Moreover, the immunoexpression of proteins related to epithelial proliferation and maturation (p63, CK10, CK14) all indicated a faster maturation of the migrating KCs in the LLLT-treated wounds. In that way, an improved epithelial healing was promoted by LLLT with the employed parameters; this improvement was confirmed by changes in the expression of several proteins related to epithelial proliferation and maturation. Immunofluorescent expression of cytokeratin 10 (red) and Cyclin D1 (green) in (A) Control keratinocytes and (B) Low-level laser irradiated cells. Blue color illustrates the nuclei of the cells (DAPI staining).


Subject(s)
Keratinocytes/cytology , Keratinocytes/radiation effects , Low-Level Light Therapy , Wound Healing/radiation effects , Animals , Cell Differentiation/radiation effects , Cell Line , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Epithelium/radiation effects , Female , Humans , Keratinocytes/metabolism , Keratins/metabolism , Rats , Rats, Wistar
2.
FEBS Open Bio ; 4: 96-104, 2014.
Article in English | MEDLINE | ID: mdl-24490130

ABSTRACT

Cisplatin-based chemotherapy is the standard treatment of choice for head and neck squamous cell carcinoma (HNSCC). The efficiency of platinum-based therapies is directly influenced by the development of tumor resistance. Multiple signaling pathways have been linked to tumor resistance, including activation of nuclear factor kappa B (NFκB). We explore a novel mechanism by which NFκB drives HNSCC resistance through histone modifications. Post-translational modification of histones alters chromatin structure, facilitating the binding of nuclear factors that mediate DNA repair, transcription, and other processes. We found that chemoresistant HNSCC cells with active NFκB signaling respond to chemotherapy by reducing nuclear BRCA1 levels and by promoting histone deacetylation (chromatin compaction). Activation of this molecular signature resulted in impaired DNA damage repair, prolonged accumulation of histone γH2AX and increased genomic instability. We found that pharmacological induction of histone acetylation using HDAC inhibitors prevented NFκB-induced cisplatin resistance. Furthermore, silencing NFκB in HNSCC induced acetylation of tumor histones, resulting in reduced chemoresistance and increased cytotoxicity following cisplatin treatment. Collectively, these findings suggest that epigenetic modifications of HNSCC resulting from NFκB-induced histone modifications constitute a novel molecular mechanism responsible for chemoresistance in HNSCC. Therefore, targeted inhibition of HDAC may be used as a viable therapeutic strategy for disrupting tumor resistance caused by NFκB.

3.
J Biophotonics ; 6(10): 839-47, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23554211

ABSTRACT

Low-level laser therapy (LLLT) is a non-thermal phototherapy used in several medical applications, including wound healing, reduction of pain and amelioration of oral mucositis. Nevertheless, the effects of LLLT upon cancer or dysplastic cells have been so far poorly studied. Head and neck cancer patients receiving LLLT for oral mucositis, for example, might have remaining tumor cells that could be stimulated by LLLT. This study demonstrated that LLLT (GaAlAs--660 nm or 780 nm, 40 mW, 2.05, 3.07 or 6.15 J/cm²) can modify oral dysplastic cells (DOK) and oral cancer cells (SCC9 and SCC25) growth by modulating the Akt/mTOR/CyclinD1 signaling pathway; LLLT significantly modified the expression of proteins related to progression and invasion in all the cell lines, and could aggravate oral cancer cellular behavior, increasing the expression of pAkt, pS6 and Cyclin D1 proteins and producing an aggressive Hsp90 isoform. Apoptosis was detected for SCC25 and was related to pAkt levels.


Subject(s)
Low-Level Light Therapy , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/radiation effects , TOR Serine-Threonine Kinases/metabolism , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Gene Expression Regulation, Enzymologic/radiation effects , Humans , Mouth Neoplasms/radiotherapy , Proto-Oncogene Mas
4.
PLoS One ; 8(3): e58672, 2013.
Article in English | MEDLINE | ID: mdl-23527004

ABSTRACT

The genome is organized and packed into the nucleus through interactions with core histone proteins. Emerging evidence suggests that tumors are highly responsive to epigenetic alterations that induce chromatin-based events and dynamically influence tumor behavior. We examined chromatin organization in head and neck squamous cell carcinoma (HNSCC) using acetylation levels of histone 3 as a marker of chromatin compaction. Compared to control oral keratinocytes, we found that HNSCC cells are hypoacetylated and that microenvironmental cues (e.g., microvasculature endothelial cells) induce tumor acetylation. Furthermore, we found that chemical inhibition of histone deacetylases (HDAC) reduces the number of cancer stem cells (CSC) and inhibits clonogenic sphere formation. Paradoxically, inhibition of HDAC also induced epithelial-mesenchymal transition (EMT) in HNSCC cells, accumulation of BMI-1, an oncogene associated with tumor aggressiveness, and expression of the vimentin mesenchymal marker. Importantly, we observed co-expression of vimentin and acetylated histone 3 at the invasion front of human HNSCC tumor tissues. Collectively, these findings suggest that environmental cues, such as endothelial cell-secreted factors, modulate tumor plasticity by limiting the population of CSC and inducing EMT. Therefore, inhibition of HDAC may constitute a novel strategy to disrupt the population of CSC in head and neck tumors to create a homogeneous population of cancer cells with biologically defined signatures and predictable behavior.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Histone Deacetylase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Acetylation , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic/drug effects , Head and Neck Neoplasms/genetics , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment/drug effects
5.
J Carcinog Mutagen ; Suppl 52013 Aug 02.
Article in English | MEDLINE | ID: mdl-25126449

ABSTRACT

Studies attempting to identify and understand the function of mutated genes and deregulated molecular pathways in cancer have been ongoing for many years. The PI3K-PTEN-mTOR signaling pathway is one of the most frequently deregulated pathways in cancer. PIK3CA mutations are found 11%-33% of head and neck cancer (HNC). The hotspot mutation sites for PIK3CA are E542K, E545K and H1047R/L. The PTEN somatic mutations are in 9-23% of HNC, and they frequently cluster in the phosphatase domain of PTEN protein. PTEN loss of heterozygosity (LOH) ranges from 41%-71% and loss of PTEN protein expression occurs in 31.2% of the HNC samples. PIK3CA and PTEN are key molecules in the PI3K-PTEN-mTOR signaling pathway. In this review, we provided a comprehensive overview of mutations in the PI3K-PTEN-mTOR molecular circuitry in HNC, including PI3K family members, TSC1/TSC2, PTEN, AKT, and mTORC1 and mTORC2 complexes. We discussed how these genetic alterations may affect protein structure and function. We also highlight the latest discoveries in protein kinase and tumor suppressor families, emphasizing how mutations in these families interfere with PI3K signaling. A better understanding of the mechanisms underlying cancer formation, progression and resistance to therapy will inform selection of novel genomic-based personalized therapies for head and neck cancer patients.

6.
Histopathology ; 60(4): 609-16, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22260414

ABSTRACT

AIMS: This study aimed to describe the expression of oestrogen receptor (ER)α, ERß and aromatase in salivary gland adenoid cystic carcinoma (ACC). METHODS AND RESULTS: ERα, ERß and aromatase expression was analysed by immunohistochemistry in tissue microarray blocks from 38 cases of ACC and seven normal salivary glands. The intracellular localization and amount of total protein expression were investigated by immunofluorescence and western blotting in an ACC cell line. Western blotting analysis showed overexpression of ERα, ERß and aromatase in the ACC cell line; however, with immunofluorescence, only ERß was shown to be expressed in the nucleus. Immunohistochemistry revealed positive nuclear expression of ERß, positive cytoplasmic expression of aromatase and a lack of ERα expression as compared with normal salivary glands. CONCLUSIONS: The nuclear expression of ERß indicates that oestrogen may be active in ACC and possibly able to mediate E2-targeted gene transcription. This study strongly suggests that ERß may be involved in tumour progression, playing a role in tumour development, and thus corroborating the indication for ER antagonists in the clinical control of ACC. This study opens a new perspective on the potential use of anti-oestrogens and aromatase inhibitors as therapeutic agents against ACC.


Subject(s)
Carcinoma, Adenoid Cystic/metabolism , Estrogen Receptor beta/metabolism , Salivary Gland Neoplasms/metabolism , Salivary Glands/metabolism , Aromatase/metabolism , Carcinoma, Adenoid Cystic/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Estrogen Receptor alpha/metabolism , Female , Humans , Male , Salivary Gland Neoplasms/pathology , Salivary Glands/pathology , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL