Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Br J Cancer ; 131(2): 220-230, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643337

ABSTRACT

The World Health Organisation recognised human papillomavirus (HPV) as the cause of multiple cancers, including head and neck cancers. HPV is a double-stranded DNA virus, and its viral gene expression can be controlled after infection by cellular and viral promoters. In cancer cells, the HPV genome is detected as either integrated into the host genome, episomal (extrachromosomal), or a mixture of integrated and episomal. Viral integration requires the breakage of both viral and host DNA, and the integration rate correlates with the level of DNA damage. Interestingly, patients with HPV-positive head and neck cancers generally have a good prognosis except for a group of patients with fully integrated HPV who show worst clinical outcomes. Those patients present with lowered expression of viral genes and limited infiltration of cytotoxic T cells. An impediment to effective therapy applications in the clinic is the sole testing for HPV positivity without considering the HPV integration status. This review will discuss HPV integration as a potential determinant of response to therapies in head and neck cancers and highlight to the field a novel therapeutic avenue that would reduce the cancer burden and improve patient survival.


Subject(s)
Head and Neck Neoplasms , Papillomavirus Infections , Humans , Head and Neck Neoplasms/virology , Head and Neck Neoplasms/therapy , Papillomavirus Infections/virology , Papillomavirus Infections/complications , Papillomaviridae/genetics , Virus Integration
2.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34180969

ABSTRACT

Ets homologous factor (EHF) is a member of the epithelial-specific Ets (ESE) family of transcription factors. To investigate its role in development and epithelial homeostasis, we generated a series of novel mouse strains in which the Ets DNA-binding domain of Ehf was deleted in all tissues (Ehf-/-) or specifically in the gut epithelium. Ehf-/- mice were born at the expected Mendelian ratio, but showed reduced body weight gain, and developed a series of pathologies requiring most Ehf-/- mice to reach an ethical endpoint before reaching 1 year of age. These included papillomas in the facial skin, abscesses in the preputial glands (males) or vulvae (females), and corneal ulcers. Ehf-/-mice also displayed increased susceptibility to experimentally induced colitis, which was confirmed in intestinal-specific Ehf knockout mice. Gut-specific Ehf deletion also impaired goblet cell differentiation, induced extensive transcriptional reprogramming in the colonic epithelium and enhanced Apc-initiated adenoma development. The Ets DNA-binding domain of EHF is therefore essential for postnatal homeostasis of the epidermis and colonic epithelium, and its loss promotes colonic tumour development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/etiology , Epidermis/metabolism , Genes, APC , Homeostasis , Intestinal Mucosa/metabolism , Transcription Factors/genetics , Animals , Cellular Reprogramming/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Gene Expression Regulation , Goblet Cells/metabolism , Goblet Cells/pathology , Male , Mice , Mice, Knockout , Transcription Factors/metabolism
3.
Dev Dyn ; 252(5): 647-667, 2023 05.
Article in English | MEDLINE | ID: mdl-36606449

ABSTRACT

BACKGROUND: The gene encoding the transcription factor, Grainyhead-like 3 (Grhl3), plays critical roles in mammalian development and homeostasis. Grhl3-null embryos exhibit thoraco-lumbo-sacral spina bifida and soft-tissue syndactyly. Additional studies reveal that these embryos also exhibit an epidermal proliferation/differentiation imbalance. This manifests as skin barrier defects resulting in peri-natal lethality and defective wound repair. Despite these extensive analyses of Grhl3 loss-of-function models, the consequences of gain-of-function of this gene have been difficult to achieve. RESULTS: In this study, we generated a novel mouse model that expresses Grhl3 from a transgene integrated in the Rosa26 locus on an endogenous Grhl3-null background. Expression of the transgene rescues both the neurulation and skin barrier defects of the knockout mice, allowing survival into adulthood. Despite this, the mice are not normal, exhibiting a range of phenotypes attributable to dysregulated Grhl3 expression. In mice homozygous for the transgene, we observe a severe Shaker-Waltzer phenotype associated with hearing impairment. Micro-CT scanning of the inner ear revealed profound structural alterations underlying these phenotypes. In addition, these mice exhibit other developmental anomalies including hair loss, digit defects, and epidermal dysmorphogenesis. CONCLUSION: Taken together, these findings indicate that diverse developmental processes display low tolerance to dysregulation of Grhl3.


Subject(s)
DNA-Binding Proteins , Spinal Dysraphism , Mice , Animals , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Spinal Dysraphism/genetics , Epidermis/metabolism , Mice, Knockout , Mammals/metabolism
4.
Mol Ther ; 29(8): 2571-2582, 2021 08 04.
Article in English | MEDLINE | ID: mdl-33775911

ABSTRACT

Current therapies for treating heterogeneous cancers such as head and neck squamous cell carcinoma (HNSCC) are non-selective and are administered independent of response biomarkers. Therapy resistance subsequently emerges, resulting in increased cellular proliferation that is associated with loss of differentiation. Whether a cancer cell differentiation potential can dictate therapy responsiveness is still currently unknown. A multi-omic approach integrating whole-genome and whole-transcriptome sequencing with drug sensitivity was employed in a HNSCC mouse model, primary patients' data, and human cell lines to assess the potential of functional differentiation in predicting therapy response. Interestingly, a subset of HNSCC with effective GRHL3-dependent differentiation was the most sensitive to inhibitors of PI3K/mTOR, c-Myc, and STAT3 signaling. Furthermore, we identified the GRHL3-differentiation target gene Filaggrin (FLG) as a response biomarker and more importantly, stratified HNSCC subsets as treatment resistant based on their FLG mutational profile. The loss of FLG in sensitive HNSCC resulted in a dramatic resistance to targeted therapies while the GRHL3-FLG signature predicted a favorable patient prognosis. This study provides evidence for a functional GRHL3-FLG tumor-specific differentiation axis that regulates targeted therapy response in HNSCC and establishes a rationale for clinical investigation of differentiation-paired targeted therapy in heterogeneous cancers.


Subject(s)
Biomarkers, Tumor/genetics , DNA-Binding Proteins/genetics , Filaggrin Proteins/genetics , Head and Neck Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Prognosis , Signal Transduction , Exome Sequencing , Whole Genome Sequencing
5.
Int J Mol Sci ; 23(5)2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35269877

ABSTRACT

Grainyhead-like (GRHL) factors are essential, highly conserved transcription factors (TFs) that regulate processes common to both natural cellular behaviours during embryogenesis, and de-regulation of growth and survival pathways in cancer. Serving to drive the transcription, and therefore activation of multiple co-ordinating pathways, the three GRHL family members (GRHL1-3) are a critical conduit for modulating the molecular landscape that guides cellular decision-making processes during proliferation, epithelial-mesenchymal transition (EMT) and migration. Animal models and in vitro approaches harbouring GRHL loss or gain-of-function are key research tools to understanding gene function, which gives confidence that resultant phenotypes and cellular behaviours may be translatable to humans. Critically, identifying and characterising the target genes to which these factors bind is also essential, as they allow us to discover and understand novel genetic pathways that could ultimately be used as targets for disease diagnosis, drug discovery and therapeutic strategies. GRHL1-3 and their transcriptional targets have been shown to drive comparable cellular processes in Drosophila, C. elegans, zebrafish and mice, and have recently also been implicated in the aetiology and/or progression of a number of human congenital disorders and cancers of epithelial origin. In this review, we will summarise the state of knowledge pertaining to the role of the GRHL family target genes in both development and cancer, primarily through understanding the genetic pathways transcriptionally regulated by these factors across disparate disease contexts.


Subject(s)
DNA-Binding Proteins , Neoplasms , Repressor Proteins/metabolism , Animals , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Epithelial-Mesenchymal Transition/genetics , Mice , Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism
6.
Int J Mol Sci ; 22(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34445392

ABSTRACT

Oral cancer is a major global health problem with high incidence and low survival rates. The oral cavity contains biofilms as dental plaques that harbour both Gram-negative and Gram-positive bacterial antigens, lipopolysaccharide (LPS) and lipoteichoic acid (LTA), respectively. LPS and LTA are known to stimulate cancer cell growth, and the bioactive phytochemical capsaicin has been reported to reverse this effect. Here, we tested the efficacy of oral cancer chemotherapy treatment with capsaicin in the presence of LPS, LTA or the combination of both antigens. LPS and LTA were administered to Cal 27 oral cancer cells prior to and/or concurrently with capsaicin, and the treatment efficacy was evaluated by measuring cell proliferation and apoptotic cell death. We found that while capsaicin inhibits oral cancer cell proliferation and metabolism (MT Glo assay) and increases cell death (Trypan blue exclusion assay and Caspase 3/7 expression), its anti-cancer effect was significantly reduced on cells that are either primed or exposed to the bacterial antigens. Capsaicin treatment significantly increased oral cancer cells' suppressor of cytokine signalling 3 gene expression. This increase was reversed in the presence of bacterial antigens during treatment. Our data establish a rationale for clinical consideration of bacterial antigens that may interfere with the treatment efficacy of oral cancer.


Subject(s)
Antigens, Bacterial/adverse effects , Capsaicin/pharmacology , Mouth Neoplasms/metabolism , Signal Transduction/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lipopolysaccharides/adverse effects , Mouth Neoplasms/drug therapy , Mouth Neoplasms/microbiology , Teichoic Acids/adverse effects
7.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830441

ABSTRACT

The bacterial antigen, lipopolysaccharide (LPS) and disruptions in calcium channels are independently known to influence oral cancer progression. Previously, we found that bacterial antigens, LPS and lipoteichoic acid (LTA) act as confounders during the action of capsaicin on Cal 27 oral cancer proliferation. As calcium channel drugs may affect oral cancer cell proliferation, we investigated the effect of ML218 HCl, a T-type voltage-gated calcium channel blocker, on the proliferation of Cal 27 oral cancer cells. We hypothesized that ML218 HCl could effectively reduce LPS-induced oral cancer cell proliferation. LPS and LTA antigens were added to Cal 27 oral cancer cells either prior to and/or concurrently with ML218 HCl treatment, and the efficacy of the treatment was evaluated by measuring Cal 27 proliferation, cell death and apoptosis. ML218 HCl inhibited oral cancer cell proliferation, increased apoptosis and cell death, but their efficacy was significantly reduced in the presence of bacterial antigens. ML218 HCl proved more effective than capsaicin in reducing bacterial antigen-induced Cal 27 oral cancer cell proliferation. Our results also suggest an interplay of proliferation factors during the bacterial antigens and calcium channel drug interaction in Cal 27. Bacterial antigen reduction of drug efficacy should be considered for developing newer pharmacological agents or testing the efficacy of the existing oral cancer chemotherapeutic agents. Finally, voltage gated calcium channel drugs should be considered for future oral cancer research.


Subject(s)
Antigens, Bacterial/genetics , Azabicyclo Compounds/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Mouth Neoplasms/drug therapy , Antigens, Bacterial/immunology , Apoptosis/drug effects , Capsaicin/pharmacology , Cell Line, Tumor , Humans , Lipopolysaccharides/toxicity , Mouth Neoplasms/chemically induced , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology
8.
Int J Mol Sci ; 20(9)2019 May 05.
Article in English | MEDLINE | ID: mdl-31060263

ABSTRACT

Squamous cell carcinomas (SCC), including cutaneous SCCs, are by far the most frequent cancers in humans, accounting for 80% of all newly diagnosed malignancies worldwide. The old dogma that SCC develops exclusively from stem cells (SC) has now changed to include progenitors, transit-amplifying and differentiated short-lived cells. Accumulation of specific oncogenic mutations is required to induce SCC from each cell population. Whilst as fewer as one genetic hit is sufficient to induce SCC from a SC, multiple events are additionally required in more differentiated cells. Interestingly, the level of differentiation correlates with the number of transforming events required to induce a stem-like phenotype, a long-lived potential and a tumourigenic capacity in a progenitor, a transient amplifying or even in a terminally differentiated cell. Furthermore, it is well described that SCCs originating from different cells of origin differ not only in their squamous differentiation status but also in their malignant characteristics. This review summarises recent findings in cutaneous SCC and highlights transforming oncogenic events in specific cell populations. It underlines oncogenes that are restricted either to stem or differentiated cells, which could provide therapeutic target selectivity against heterogeneous SCC. This strategy may be applicable to SCC from different body locations, such as head and neck SCCs, which are currently still associated with poor survival outcomes.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/etiology , Neoplastic Stem Cells/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/etiology , Animals , Biomarkers, Tumor , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic/drug effects , Genome-Wide Association Study , Humans , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Skin Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
Dev Biol ; 429(1): 12-19, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28629792

ABSTRACT

The two main mechanisms that expand the proteomic output of eukaryotic genes are alternative splicing and alternative translation initiation signals. Despite being essential to generate isoforms of gene products that create functional diversity during development, the impact of these mechanisms on fine-tuning regulatory gene networks is still underappreciated. In this review, we use the Grainyhead-like (Grhl) family as a case study to illustrate the importance of isoforms when investigating transcription factor family function during development and disease, and highlight the potential for differential modulation of downstream target genes. We provide insights into the importance of considering alternative gene products when designing, undertaking, and analysing primary research, and the effect that isoforms may have on development. This review also covers known mutations in Grhl family members, and postulates how genetic changes may dictate transcriptional specificity between the Grhl family members. It also contrasts and compares the available literature on the function and importance of the Grhl isoforms, and highlights current gaps in our understanding of their regulatory gene networks in development and disease.


Subject(s)
Alternative Splicing/genetics , Transcription Factors/metabolism , Transcription Initiation Site , Animals , Humans , Mutation/genetics , Protein Domains , Protein Processing, Post-Translational/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
10.
Int J Mol Sci ; 18(7)2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28654000

ABSTRACT

Non-melanomatous skin cancers (NMSCs), which include basal and squamous cell carcinoma (BCC and SCC respectively), represent a significant burden on the population, as well as an economic load to the health care system; yet treatments of these preventable cancers remain ineffective. Studies estimate that there has been a 2-fold increase in the incidence of NMSCs between the 1960s and 1980s. The increase in cases of NMSCs, as well as the lack of effective treatments, makes the need for novel therapeutic approaches all the more necessary. To rationally develop more targeted treatments for NMSCs, a better understanding of the cell of origin, in addition to the underlying pathophysiological mechanisms that govern the development of these cancers, is urgently required. Research over the past few years has provided data supporting both a "bottom up" and "top down" mechanism of tumourigenesis. The "bottom up" concept involves a cancer stem cell originating in the basal compartment of the skin, which ordinarily houses the progenitor cells that contribute towards wound healing and normal cell turnover of overlying epidermal skin layers. The "top down" concept involves a more differentiated cell undergoing genetic modifications leading to dedifferentiation, giving rise to cancer initiating cells (CICs). This review explores both concepts, to paint a picture of the skin SCC cell of origin, the underlying biology, and also how this knowledge might be exploited to develop novel therapies.


Subject(s)
Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Epidermis/pathology , Neoplastic Stem Cells/pathology , Skin Neoplasms/pathology , Skin/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Basal Cell/genetics , Carcinoma, Squamous Cell/genetics , Cell Dedifferentiation , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Epidermis/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/metabolism , Skin/metabolism , Skin Neoplasms/genetics , Transcription Factors/analysis , Transcription Factors/genetics
11.
Development ; 139(3): 525-36, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22223680

ABSTRACT

The isthmic organiser located at the midbrain-hindbrain boundary (MHB) is the crucial developmental signalling centre responsible for patterning mesencephalic and metencephalic regions of the vertebrate brain. Formation and maintenance of the MHB is characterised by a hierarchical program of gene expression initiated by fibroblast growth factor 8 (Fgf8), coupled with cellular morphogenesis, culminating in the formation of the tectal-isthmo-cerebellar structures. Here, we show in zebrafish that one orthologue of the transcription factor grainy head-like 2 (Grhl2), zebrafish grhl2b plays a central role in both MHB maintenance and folding by regulating two distinct, non-linear pathways. Loss of grhl2b expression induces neural apoptosis and extinction of MHB markers, which are rescued by re-expression of engrailed 2a (eng2a), an evolutionarily conserved target of the Grhl family. Co-injection of sub-phenotypic doses of grhl2b and eng2a morpholinos reproduces the apoptosis and MHB marker loss, but fails to substantially disrupt formation of the isthmic constriction. By contrast, a novel direct grhl2b target, spec1, identified by phylogenetic analysis and confirmed by ChIP, functionally cooperates with grhl2b to induce MHB morphogenesis, but plays no role in apoptosis or maintenance of MHB markers. Collectively, these data show that MHB maintenance and morphogenesis are dissociable events regulated by grhl2b through diverse transcriptional targets.


Subject(s)
Carrier Proteins/metabolism , Mesencephalon/growth & development , Morphogenesis , Rhombencephalon/growth & development , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , Apoptosis , Carrier Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mesencephalon/metabolism , Morpholinos/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Phylogeny , Rhombencephalon/metabolism , Signal Transduction , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 107(6): 2628-33, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133805

ABSTRACT

Symplekin is a ubiquitously expressed protein involved in cytoplasmic RNA polyadenylation and transcriptional regulation and is localized at tight junctions (TJs) in epithelial cells. Nuclear symplekin cooperates with the Y-box transcription factor zonula occludens 1-associated nucleic acid-binding protein (ZONAB) to increase the transcription of cell cycle-related genes and also inhibits differentiation of intestinal cells. We detected high levels of nuclear symplekin in 8 of 12 human colorectal cancer (CRC) samples. shRNA-mediated reduction of symplekin expression was sufficient to decrease significantly the anchorage-independent growth and proliferation of HT-29 CRC cells as well as their tumorigenicity when injected into immunodeficient animals. Symplekin down-regulation also was found to alter ion transport through TJs, to promote the localization of ZONAB in the membrane rather than the nucleus, and strongly to enhance cell polarization in a 3D matrix, leading to the formation of spheroids organized around a central lumen. Claudin-2 expression was reduced following symplekin down-regulation, an effect mimicked when ZONAB expression was down-regulated using selective siRNA. Virus-mediated restoration of claudin-2 expression was found to restore nuclear expression of ZONAB in HT29DeltaSym cells and to rescue the phenotypic alterations induced by symplekin down-regulation of cell polarity, paracellular transport, ZONAB localization, cyclin D1 expression, proliferation, and anchorage-independent growth. Finally, siRNA-mediated claudin-2 down-regulation increased the transepithelial resistance and decreased cyclin D1 expression and ZONAB nuclear localization, similar to observations in symplekin-depleted cells. Our results suggest that nuclear overexpression of symplekin promotes tumorigenesis in the human colon and that the regulation of claudin-2 expression is instrumental in this effect.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nuclear Proteins/genetics , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Proliferation , Claudins , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Fluorescent Antibody Technique , HT29 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden , Up-Regulation
13.
Int J Mol Sci ; 14(7): 13670-86, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23812084

ABSTRACT

Epidermal stem cells sustain the adult skin for a lifetime through self-renewal and the production of committed progenitors. These stem cells generate progeny that will undergo terminal differentiation leading to the development of a protective epidermal barrier. Whereas the molecular mechanisms that govern epidermal barrier repair and renewal have been extensively studied, pathways controlling stem cell differentiation remain poorly understood. Asymmetric cell divisions, small non-coding RNAs (microRNAs), chromatin remodeling complexes, and multiple differentiation factors tightly control the balance of stem and progenitor cell proliferation and differentiation, and disruption of this balance leads to skin diseases. In this review, we summarize and discuss current advances in our understanding of the mechanisms regulating epidermal stem and progenitor cell differentiation, and explore new relationships for maintenance of skin barrier function.


Subject(s)
Adult Stem Cells , Cell Differentiation/physiology , Cell Division/physiology , Cell Proliferation/physiology , Chromatin Assembly and Disassembly/physiology , Epidermis , Adult , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Epidermal Cells , Epidermis/metabolism , Humans , MicroRNAs/metabolism
14.
Cancers (Basel) ; 15(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36765809

ABSTRACT

Breakthrough research in the field of immune checkpoint inhibitors and the development of a human papilloma virus vaccine triggered a plethora of research in the field of cancer immunotherapy. Both had significant effects on the treatment of head and neck squamous cell carcinoma. The advent of preclinical models and multidisciplinary approaches including bioinformatics, genetic engineering, clinical oncology, and immunology helped in the development of tumour-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy. Here, we discuss different immunotherapies such as adoptive T-cell transfer, immune checkpoint inhibitors, interleukins, and cancer vaccines for the treatment of head and neck cancer. This review showcases the intrinsic relation between the understanding and implementation of basic biology and clinical practice. We also address potential limitations of each immunotherapy approach and the advantages of personalized immunotherapy. Overall, the aim of this review is to encourage further research in the field of immunotherapy for head and neck cancer.

15.
Sci Rep ; 13(1): 17033, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37813936

ABSTRACT

The tumour-cell based initiation of immune evasion project evaluated the role of Gipie in adenoid cystic carcinoma (ACC) and mucoepidermoid carcinoma (A-253), from ninety-six 3D-ACC and A-253-immune co-culture models using natural killer cells (NK), and Jurkat cells (JK). Abnormal ACC morphology was observed in 3D-ACC immune co-culture models. Gipie-silencing conferred a "lymphoblast-like" morphology to ACC cells, a six-fold increase in apoptotic cells (compared to unaltered ACC cells, P ≤ 0.0001), a two-fold decrease in T regulatory cells (FoxP3+/IL-2Rα+/CD25+) (P ≤ 0.0001), and a three-fold increase in activated NK cells (NKp30+/IFN-γ+) (P ≤ 0.0001) with significantly higher release of granzyme (P ≤ 0.001) and perforin (P ≤ 0.0001).


Subject(s)
Carcinoma, Adenoid Cystic , Humans , Carcinoma, Adenoid Cystic/pathology , Killer Cells, Natural , T-Lymphocytes, Regulatory , Jurkat Cells , Perforin
16.
Nat Commun ; 14(1): 1591, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949044

ABSTRACT

In heterogeneous head and neck cancer (HNC), subtype-specific treatment regimens are currently missing. An integrated analysis of patient HNC subtypes using single-cell sequencing and proteome profiles reveals an epithelial-mesenchymal transition (EMT) signature within the epithelial cancer-cell population. The EMT signature coincides with PI3K/mTOR inactivation in the mesenchymal subtype. Conversely, the signature is suppressed in epithelial cells of the basal subtype which exhibits hyperactive PI3K/mTOR signalling. We further identify YBX1 phosphorylation, downstream of the PI3K/mTOR pathway, restraining basal-like cancer cell proliferation. In contrast, YBX1 acts as a safeguard against the proliferation-to-invasion switch in mesenchymal-like epithelial cancer cells, and its loss accentuates partial-EMT and in vivo invasion. Interestingly, phospho-YBX1 that is mutually exclusive to partial-EMT, emerges as a prognostic marker for overall patient outcomes. These findings create a unique opportunity to sensitise mesenchymal cancer cells to PI3K/mTOR inhibitors by shifting them towards a basal-like subtype as a promising therapeutic approach against HNC.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Cell Proliferation/genetics , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Cell Movement , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
17.
Dev Biol ; 349(2): 512-22, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21081122

ABSTRACT

The Grainy head-like 3 (Grhl3) gene encodes a transcription factor that plays essential roles in epidermal morphogenesis during embryonic development, with deficient mice exhibiting failed skin barrier formation, defective wound repair, and loss of eyelid fusion. Despite sharing significant sequence homology, overlapping expression patterns, and an identical core consensus DNA binding site, the other members of the Grhl family (Grhl1 and -2) fail to compensate for the loss of Grhl3 in these processes. Here, we have employed diverse genetic models, coupled with biochemical studies, to define the inter-relationships of the Grhl factors in epidermal development. We show that Grhl1 and Grhl3 have evolved complete functional independence, as evidenced by a lack of genetic interactions in embryos carrying combinations of targeted alleles of these genes. In contrast, compound heterozygous Grhl2/Grhl3 embryos displayed failed wound repair, and loss of a single Grhl2 allele in Grhl3-null embryos results in fully penetrant eyes open at birth. Expression of Grhl2 from the Grhl3 locus in homozygous knock-in mice corrects the wound repair defect, but these embryos still display a complete failure of skin barrier formation. This functional dissociation is due to unexpected differences in target gene specificity, as both GRHL2 and GRHL3 bind to and regulate expression of the wound repair gene Rho GEF 19, but regulation of the barrier forming gene, Transglutaminase 1 (TGase1), is unique to GRHL3. Our findings define the mechanisms underpinning the unique and cooperative roles of the Grhl genes in epidermal development.


Subject(s)
DNA-Binding Proteins/metabolism , Epidermis/embryology , Morphogenesis/physiology , Phenotype , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , DNA Primers/genetics , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Epidermis/ultrastructure , Gene Knock-In Techniques , Immunohistochemistry , In Situ Hybridization , Mice , Microscopy, Electron, Scanning , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transglutaminases/metabolism , Wound Healing/physiology
18.
J Cell Biol ; 178(4): 635-48, 2007 Aug 13.
Article in English | MEDLINE | ID: mdl-17698607

ABSTRACT

The HMG-box transcription factor Sox9 is expressed in the intestinal epithelium, specifically, in stem/progenitor cells and in Paneth cells. Sox9 expression requires an active beta-catenin-Tcf complex, the transcriptional effector of the Wnt pathway. This pathway is critical for numerous aspects of the intestinal epithelium physiopathology, but processes that specify the cell response to such multipotential signals still remain to be identified. We inactivated the Sox9 gene in the intestinal epithelium to analyze its physiological function. Sox9 inactivation affected differentiation throughout the intestinal epithelium, with a disappearance of Paneth cells and a decrease of the goblet cell lineage. Additionally, the morphology of the colon epithelium was severely altered. We detected general hyperplasia and local crypt dysplasia in the intestine, and Wnt pathway target genes were up-regulated. These results highlight the central position of Sox9 as both a transcriptional target and a regulator of the Wnt pathway in the regulation of intestinal epithelium homeostasis.


Subject(s)
Colon/metabolism , High Mobility Group Proteins/metabolism , Paneth Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Colon/cytology , High Mobility Group Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , SOX9 Transcription Factor , Transcription Factors/genetics
19.
Cancers (Basel) ; 14(10)2022 May 20.
Article in English | MEDLINE | ID: mdl-35626124

ABSTRACT

Squamous cell carcinomas (SCCs) are cancers of epithelial cells lining the aerodigestive and genitourinary tract [...].

20.
Cancers (Basel) ; 13(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34680271

ABSTRACT

The oral epithelium is one of the fastest repairing and continuously renewing tissues. Stem cell activation within the basal layer of the oral epithelium fuels the rapid proliferation of multipotent progenitors. Stem cells first undergo asymmetric cell division that requires tightly controlled and orchestrated differentiation networks to maintain the pool of stem cells while producing progenitors fated for differentiation. Rapidly expanding progenitors subsequently commit to advanced differentiation programs towards terminal differentiation, a process that regulates the structural integrity and homeostasis of the oral epithelium. Therefore, the balance between differentiation and terminal differentiation of stem cells and their progeny ensures progenitors commitment to terminal differentiation and prevents epithelial transformation and oral squamous cell carcinoma (OSCC). A recent comprehensive molecular characterization of OSCC revealed that a disruption of terminal differentiation factors is indeed a common OSCC event and is superior to oncogenic activation. Here, we discuss the role of differentiation and terminal differentiation in maintaining oral epithelial homeostasis and define terminal differentiation as a critical tumour suppressive mechanism. We further highlight factors with crucial terminal differentiation functions and detail the underlying consequences of their loss. Switching on terminal differentiation in differentiated progenitors is likely to represent an extremely promising novel avenue that may improve therapeutic interventions against OSCC.

SELECTION OF CITATIONS
SEARCH DETAIL