Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Int J Nanomedicine ; 19: 9333-9349, 2024.
Article in English | MEDLINE | ID: mdl-39286354

ABSTRACT

Introduction: Immunotherapy has led to a paradigm shift in reinvigorating treatment of cancer. Nevertheless, tumor associated macrophages (TAMs) experience functional polarization on account of the generation of suppressive metabolites, contributing to impaired antitumor immune responses. Methods: Hence, metabolic reprogramming of tumor microenvironment (TME) can synergistically improve the efficacy of anti-tumor immunotherapy. Herein, we engineered an iron-based nanoplatform termed ERFe3O4 NPs. This platform features hollow Fe3O4 nanoparticles loaded with the natural product emodin, the outer layer is coated with red blood cell membrane (mRBCs) inserted with DSPE-PEG2000-galactose. This effectively modulates lactate production, thereby reversing the tumor immune suppressive microenvironment (TIME). Results: The ERFe3O4 NPs actively targeted TAMs on account of their ability to bind to M2-like TAMs with high expression of galectin (Mgl). ERFe3O4 NPs achieved efficient ability to reverse TIME via the production of reducing lactate and prompting enrichment iron of high concentrations. Furthermore, ERFe3O4 NPs resulted in heightened expression of CD16/32 and enhanced TNF-α release, indicating promotion of M1 TAMs polarization. In vitro and in vivo experiments revealed that ERFe3O4 NPs induced significant apoptosis of tumor cells and antitumor immune response. Discussion: This study combines Traditional Chinese Medicine (TCM) with nanomaterials to synergistically reprogram TAMs and reverse TIME, opening up new ideas for improving anti-tumor immunotherapy.


Subject(s)
Immunotherapy , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Immunotherapy/methods , Mice , Cell Line, Tumor , Humans , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Mice, Inbred C57BL , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Apoptosis/drug effects , Iron/chemistry , Female
2.
Oral Dis ; 2023 May 15.
Article in English | MEDLINE | ID: mdl-37183989

ABSTRACT

OBJECTIVE: Oral squamous cell carcinoma (OSCC) is the most common malignant tumour in the oral cavity. OSCC is aggressive and prone to metastasis; it is associated with high mortality and short survival. In this study, we investigated the function of the long non-coding RNA LINC00525 in OSCC progression and the molecular mechanisms through in vitro and in vivo experiments. MATERIALS AND METHODS: CCK8 assay was used to detect the effect of LINC00525 on cell viability; transwell migration and invasion assays and scratch assay were used to examine the role of LINC00525 in cell migration and invasion. Flow cytometry, RT-PCR and western blot were used to detect apoptosis indexes. Tumorigenic effects were investigated using mouse xenograft tumour models. RESULTS: LINC00525 was associated with OSCC survival and prognosis. LINC00525 knockdown decreased cell viability and epithelial-mesenchymal transition (EMT) properties and increased apoptosis and also shortened the cell cycle of OSCC cells in vitro. The downregulation of LINC00525 reduced the growth of OSCC tumour in vivo. LINC00525 can regulate OSCC cells via the apoptotic signalling pathway. CONCLUSION: Our results indicate that LINC00525 exhibits oncogenic functions in OSCC. LINC00525 may be a new promising and potential target for the treatment of OSCC.

3.
PLoS Genet ; 17(2): e1009283, 2021 02.
Article in English | MEDLINE | ID: mdl-33539340

ABSTRACT

Activating transcription factor 3 (ATF3) is a key transcription factor involved in regulating cellular stress responses, with different expression levels and functions in different tissues. ATF3 has also been shown to play crucial roles in regulating tumor development and progression, however its potential role in oral squamous cell carcinomas has not been fully explored. In this study, we examined biopsies of tongue squamous cell carcinomas (TSCCs) and found that the nuclear expression level of ATF3 correlated negatively with the differentiation status of TSCCs, which was validated by analysis of the ATGC database. By using gain- or loss- of function analyses of ATF3 in four different TSCC cell lines, we demonstrated that ATF3 negatively regulates the growth and migration of human TSCC cells in vitro. RNA-seq analysis identified two new downstream targets of ATF3, interferon alpha inducible proteins 6 (IFI6) and 27 (IFI27), which were upregulated in ATF3-deleted cells and were downregulated in ATF3-overexpressing cells. Chromatin immunoprecipitation assays showed that ATF3 binds the promoter regions of the IFI6 and IFI27 genes. Both IFI6 and IFI27 were highly expressed in TSCC biopsies and knockdown of either IFI6 or IFI27 in TSCC cells blocked the cell growth and migration induced by the deletion of ATF3. Conversely, overexpression of either IFI6 or IFI27 counteracted the inhibition of TSCC cell growth and migration induced by the overexpression of ATF3. Finally, an in vivo study in mice confirmed those in vitro findings. Our study suggests that ATF3 plays an anti-tumor function in TSCCs through the negative regulation of its downstream targets, IFI6 and IFI27.


Subject(s)
Activating Transcription Factor 3/metabolism , Carcinoma, Squamous Cell/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Tongue Neoplasms/metabolism , Activating Transcription Factor 3/genetics , Animals , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Immunohistochemistry , Membrane Proteins/genetics , Mice , Mice, Nude , Mitochondrial Proteins/genetics , Neoplasm Grading , Promoter Regions, Genetic , RNA, Small Interfering , RNA-Seq , Tongue Neoplasms/genetics , Tongue Neoplasms/pathology , Up-Regulation
4.
Lab Invest ; 101(5): 636-647, 2021 05.
Article in English | MEDLINE | ID: mdl-33299127

ABSTRACT

Activating transcription factor 3 (ATF-3), a cyclic AMP-dependent transcription factor, has been shown to play a regulatory role in melanoma, although its function during tumor progression remains unclear. Here, we demonstrate that ATF-3 exhibits tumor suppressive function in melanoma. Specifically, ATF-3 nuclear expression was significantly diminished with melanoma progression from nevi to primary to metastatic patient melanomas, correlating low expression with poor prognosis. Significantly low expression of ATF-3 was also found in cultured human metastatic melanoma cell lines. Importantly, overexpression of ATF-3 in metastatic melanoma cell lines significantly inhibited cell growth, migration, and invasion in vitro; as well as abrogated tumor growth in a human melanoma xenograft mouse model in vivo. RNA sequencing analysis revealed downregulation of ERK and AKT pathways and upregulation in apoptotic-related genes in ATF-3 overexpressed melanoma cell lines, which was further validated by Western-blot analysis. In summary, this study demonstrated that diminished ATF-3 expression is associated with melanoma virulence and thus provides a potential target for novel therapies and prognostic biomarker applications.


Subject(s)
Activating Transcription Factor 3/metabolism , Melanoma/metabolism , Animals , Apoptosis , Female , Humans , MAP Kinase Signaling System , Melanoma, Experimental/metabolism , Mice, Nude , Oncogene Protein v-akt/metabolism , Phosphorylation , Retrospective Studies
5.
Front Oncol ; 10: 624, 2020.
Article in English | MEDLINE | ID: mdl-32373541

ABSTRACT

The treatment of melanoma has remained a difficult challenge. Targeting the tumor stroma has recently attracted attention for developing novel strategies for melanoma therapy. Activating transcription factor 3 (ATF3) plays a crucial role in regulating tumorigenesis and development, but whether the expression of ATF3 in human dermal fibroblasts (HDFs) can affect melanoma development hasn't been studied. Our results show that ATF3 expression is downregulated in stromal cells of human melanoma. HDFs expressing high levels of ATF3 suppressed the growth and migration of melanoma cells in association with downregulation of different cytokines including IL-6 in vitro. In vivo, HDFs with high ATF3 expression reduced tumor formation. Adding recombinant IL-6 to melanoma cells reversed those in vitro and in vivo effects, suggesting that ATF3 expression by HDFs regulates melanoma progression through the IL-6/STAT3 pathway. More importantly, HDFs pretreated with cyclosporine A or phenformin to induce ATF3 expression inhibited melanoma cell growth in vitro and in vivo. In summary, our study reveals that ATF3 suppresses human melanoma growth and that inducing the expression of ATF3 in HDFs can inhibit melanoma growth, a new potential melanoma therapeutic approach.

6.
Pigment Cell Melanoma Res ; 33(1): 16-29, 2020 01.
Article in English | MEDLINE | ID: mdl-31386789

ABSTRACT

Primary melanocytes isolated from skin and expanded in culture have been widely used for laboratory research and clinical applications. The conventional method to isolate primary melanocytes from skin usually requires about 3-4 weeks of culture for melanocytes to grow sufficiently to passage. Considering that melanocytes comprise only 3%-7% of epidermal cells in normal human skin, it would be extremely helpful to increase the isolation efficiency and shorten the initial culture time to quickly meet various application needs. Here, we report that adding Y-27632, a Rho kinase inhibitor, into the initial culture medium for 2 days can dramatically increase the yield of melanocytes. We found that Y-27632 can promote keratinocyte attachment and survival in the melanocyte culture system, resulting in not only better recovery, but also increased proliferation of melanocytes by a paracrine signaling pathway. More specifically, Y-27632 significantly induced keratinocyte expression of stem cell factor, which played an important role in enhancing the growth of melanocytes. In summary, Y-27632 could profoundly enhance the yield of primary melanocytes in the initial culture through paracrine effects on keratinocytes.


Subject(s)
Keratinocytes/cytology , Keratinocytes/metabolism , Melanocytes/metabolism , Paracrine Communication , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Culture Media , Humans , Keratinocytes/drug effects , Male , Melanocytes/drug effects , Paracrine Communication/drug effects , Pyridines/pharmacology , Signal Transduction/drug effects , Stem Cell Factor/metabolism , rho-Associated Kinases/metabolism
7.
J Invest Dermatol ; 140(6): 1266-1275.e3, 2020 06.
Article in English | MEDLINE | ID: mdl-31837302

ABSTRACT

Epigenetic regulation has a profound influence on stem cell fate during normal development in maintenance of physiologic tissue homeostasis. Here we report diminished ten-eleven translocation (TET) methylcytosine dioxygenase expression and loss of the DNA hydroxymethylation mark 5-hydroxymethylcytosine (5-hmC) in keratinocyte stem cells and transit amplifying cells in human psoriasis and in imiquimod-induced murine psoriasis. Loss of 5-hmC was associated with dysregulated keratinocyte stem cell kinetics, resulting in accumulation of nestin and FABP5-expressing transit amplifying cells to produce classic psoriatic epidermal architecture. Moreover, 5-hmC loss was accompanied by diminished TET1 and TET2 mRNA expression. Genome-wide mapping of epidermal 5-hmC in murine psoriasis revealed loci-specific loss of 5-hmC in genes regulating stem cell homeostasis, including MBD1, RTN1, STRN4, PRKD2, AKT1, and MAPKAP2, as well as those associated with RAR and Wnt/ß-catenin signaling pathways. In vitro restoration of TET expression by ascorbic acid was accomplished in cultured human keratinocyte stem cells to show similar Ca++-induced differentiation, resulting in increased 5-hmC levels and reduced nestin expression. To our knowledge, an epigenetic deficiency in psoriasis with relevance to stem cell dysregulation has not been previously reported. This observation raises the possibility that epigenetic modifiers that impact on the TET-5-hmC pathway may be a relevant approach of heretofore unappreciated therapeutic utility.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Psoriasis/genetics , 5-Methylcytosine/metabolism , Animals , DNA-Binding Proteins/metabolism , Dioxygenases , Disease Models, Animal , Down-Regulation , Female , Histone Code/genetics , Humans , Keratinocytes/pathology , Mice , Mixed Function Oxygenases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins/metabolism , Psoriasis/pathology , Sequence Analysis, DNA , Stem Cells/pathology
8.
Lab Invest ; 99(8): 1193-1202, 2019 07.
Article in English | MEDLINE | ID: mdl-30837678

ABSTRACT

Hypoxia resulting in hypoxia-inducible factor-1 alpha (HIF-1α) induction is known to drive scar formation during cutaneous wound healing, and may be responsible for excessive fibrosis inherent to hypertrophic scars and keloids. Because epigenetic pathways play an important role in regulation of fibrosing processes, we evaluated patient scars for DNA hydroxymethylation (5-hydroxymethylcytosine; 5-hmC) status and documented a significant decrease in scar fibroblasts. To test this finding in vitro, human fibroblasts were cultured with cobalt chloride (CoCl2), a known stimulant of HIF-1α. HIF-1α induced so resulted in loss of 5-hmC similar to that seen in naturally occurring scars and was associated with significant downregulation of one of the 5-hmC converting enzymes-ten-eleven translocation 3 (TET3)-as well as increased expression of phosphorylated focal adhesion kinase (p-FAK), which is important in wound contracture. These changes were partially reversed by exposure to ascorbic acid, a recognized epigenetic regulator potentially capable of minimizing excessive scar formation and promoting a more regenerative healing response. Our results provide a novel and translationally relevant mechanism whereby epigenetic regulation of scar formation may be manipulated at the level of fibroblast DNA hydroxymethylation.


Subject(s)
5-Methylcytosine/analogs & derivatives , Ascorbic Acid/pharmacology , Cell Hypoxia , Dioxygenases/metabolism , Fibroblasts/metabolism , 5-Methylcytosine/metabolism , Cells, Cultured , Cicatrix/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
9.
PLoS Genet ; 14(5): e1007366, 2018 05.
Article in English | MEDLINE | ID: mdl-29723192

ABSTRACT

An increased incidence of skin inflammatory diseases is frequently observed in organtransplanted patients being treated with calcineurin inhibitor-based immunosuppressive agents. The mechanism of increased skin inflammation in this context has however not yet been clarified. Here we report an increased inflammation following inhibition of calcineurin signaling seen in both chemically induced mouse skin tumors and in tumors grafted from H-rasV12 expressing primary human keratinocytes (HKCs). Following UVB or TPA treatment, we specifically found that deletion of the calcineurin gene in mouse keratinocytes (MKCs) resulted in increased inflammation, and this was accompanied by the enhanced production of pro-inflammatory cytokines, such as TNFα, IL-8 and CXCL1. Furthermore, expression of the RNA-binding protein, tristetraprolin (TTP) was down-regulated in response to calcineurin inhibition, wherein TTP was shown to negatively regulate the production of pro-inflammatory cytokines in keratinocytes. The induction of TTP following TPA or UVB treatment was attenuated by calcineurin inhibition in keratinocytes, and correspondingly, disruption of calcineurin signaling down-regulated the amounts of TTP in both clinical and H-rasV12-transformed keratinocyte tumor models. Our results further demonstrated that calcineurin positively controls the stabilization of TTP in keratinocytes through a proteasome-dependent mechanism. Reducing the expression of TTP functionally promoted tumor growth of H-rasV12 expressing HKCs, while stabilizing TTP expression counteracted the tumor-promoting effects of calcineurin inhibition. Collectively these results suggest that calcineurin signaling, acting through TTP protein level stabilization, suppresses keratinocyte tumors by downregulating skin inflammation.


Subject(s)
Calcineurin/metabolism , Keratinocytes/metabolism , Skin/metabolism , Tristetraprolin/metabolism , Animals , Animals, Newborn , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Inflammation Mediators/metabolism , Keratinocytes/drug effects , Keratinocytes/radiation effects , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Tristetraprolin/genetics , Ultraviolet Rays
10.
J Tissue Eng Regen Med ; 12(2): e1251-e1255, 2018 02.
Article in English | MEDLINE | ID: mdl-28752652

ABSTRACT

Primary skin epidermal cells isolation and in vitro expansion culture have been widely used for laboratory research and clinical applications. The conventional methods involving sequential enzymatic digestion of adult tissues have given low cell recovery rate and reduced cell viability. We report here an advanced method for human primary epidermal progenitor cells isolation from skin tissues including the Rho kinase inhibitor Y-27632. Compared with traditional protocols, the current protocol is simple, easy, and faster; moreover, it gives a greater yield of integrin-expressing epithelial stem cells. In addition, our new methodology does not require a separation of epidermis from dermis because the medium selectively blocks focal adhesion and growth of dermal cells. Importantly, the cells isolated from this method can maintain their regeneration potential and quickly reconstitute a mature human skin in vivo after grafting onto nude mice. In brief, we describe here a simple (one step) and serum-free method for isolating primary epidermal stem cells from adult tissues. The isolated cells may be widely used for both laboratory studies and clinical application, especially in the field of tissue engineering and regeneration.


Subject(s)
Amides/pharmacology , Cell Separation/methods , Dermis/cytology , Epidermal Cells/cytology , Focal Adhesions/metabolism , Pyridines/pharmacology , Adult , Cells, Cultured , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Focal Adhesions/drug effects , Humans , Regeneration/drug effects
11.
Regen Med ; 12(5): 503-511, 2017 07.
Article in English | MEDLINE | ID: mdl-28749726

ABSTRACT

AIM: We tested whether the a simple injection known as the patch assay could reconstitute mature hair follicles by culture-expanded human cells and explored whether the assay could reflect the trichogenicity of cultured cells. MATERIALS & METHODS: Dissociated culture-expanded fetal or adult scalp dermal cells combined with foreskin keratinocytes were subcutaneously injected into the back skin of immunosuppressive mice to form the patch skin. The patches were collected and characterized and were analyzed for hair formation efficiency. RESULTS: Using culture-expanded human fetal cells, the patch assay can efficiently reconstitute mature hair follicles and the efficiency of hair formation in the patch assay correlates with cell trichogenicity. CONCLUSION: The patch assay has the potential for testing the trichogenicity of human cells.


Subject(s)
Cell Culture Techniques/methods , Hair Follicle/cytology , Animals , Cell Proliferation , Cells, Cultured , Humans , Injections, Subcutaneous , Male , Mice, Nude
12.
Shanghai Kou Qiang Yi Xue ; 20(4): 413-6, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-21909610

ABSTRACT

PURPOSE: The purpose of this study was to investigate the somatic mutations of human mitochondria succinate dehydrogenase subunit B (SDHB) in sporadic paragangliomas. METHODS: Eight exons of SDHB gene in 8 sporadic paragangliomas cases were amplified by PCR and sequenced, respectively. The sequences were analyzed to find mutations compared with human homology sequence in Genebank and SNP database. RESULTS: Nine sequence variations were found in 8 cases, in which one mutation was found in one case (1/8, 12.5%). The mutation was identified as the sixty four base pair in exon 2 of SDHB(c.136C>T), resulting in a change from a arginine to a stop codon (p.Arg90X). The left 8 variations were polymorphisms. CONCLUSIONS: The mutation of SDHB exists in sporadic paragangliomas patients and it might play a significant role in paragangliomas tumorigenesis.


Subject(s)
Germ-Line Mutation , Succinic Acid , DNA Mutational Analysis , Humans , Mutation , Paraganglioma , Succinate Dehydrogenase , Succinates
SELECTION OF CITATIONS
SEARCH DETAIL