Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(7)2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33805154

ABSTRACT

Intermediate junctional epidermolysis bullosa caused by mutations in the COL17A1 gene is characterized by the frequent development of blisters and erosions on the skin and mucous membranes. The rarity of the disease and the heterogeneity of the underlying mutations renders therapy developments challenging. However, the high number of short in-frame exons facilitates the use of antisense oligonucleotides (AON) to restore collagen 17 (C17) expression by inducing exon skipping. In a personalized approach, we designed and tested three AONs in combination with a cationic liposomal carrier for their ability to induce skipping of COL17A1 exon 7 in 2D culture and in 3D skin equivalents. We show that AON-induced exon skipping excludes the targeted exon from pre-mRNA processing, which restores the reading frame, leading to the expression of a slightly truncated protein. Furthermore, the expression and correct deposition of C17 at the dermal-epidermal junction indicates its functionality. Thus, we assume AON-mediated exon skipping to be a promising tool for the treatment of junctional epidermolysis bullosa, particularly applicable in a personalized manner for rare genotypes.


Subject(s)
Autoantigens/metabolism , Epidermolysis Bullosa, Junctional/genetics , Non-Fibrillar Collagens/metabolism , Oligonucleotides, Antisense/genetics , RNA Splicing , Alternative Splicing , Biopsy , Cell Line , Cell Survival , Epidermolysis Bullosa, Junctional/metabolism , Epidermolysis Bullosa, Junctional/therapy , Exons , Genotype , Homozygote , Humans , Keratinocytes/cytology , Liposomes/chemistry , Mutation , Organ Culture Techniques , RNA, Messenger/metabolism , Collagen Type XVII
2.
Int J Radiat Oncol Biol Phys ; 100(4): 906-915, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29485070

ABSTRACT

PURPOSE: To report the results of a phase 1 trial evaluating the safety of the ipilimumab/radiation therapy combination in patients with metastatic melanoma. PATIENTS AND METHODS: Thirteen patients with metastatic melanoma were enrolled. Trial treatment consisted of 4 cycles of ipilimumab in combination with concurrent dose-escalated high-dose radiation therapy to 1 lesion administered before the third cycle of ipilimumab. RESULTS: Grade 3 or 4 ipilimumab-related adverse events occurred in 25% of patients. The maximum tolerated radiation therapy dose was not reached. Local control of the irradiated lesions was achieved in 11 of 12 irradiated patients (1 patient had progressive disease before irradiation and dropped out of the trial). Evaluation of the nonirradiated lesions demonstrated that 3 of 13 patients experienced clinical benefit, with 1 patient developing a partial response and 2 patients having confirmed stable disease. Immunomonitoring data showed that in patients without clinical benefit, factors linked to immunotolerance increased early after the initiation of ipilimumab, suggesting that early initiation of radiation therapy might be more effective if combined with ipilimumab. CONCLUSIONS: Our findings suggest that the combination of ipilimumab and high-dose radiation therapy is feasible and safe.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Ipilimumab/administration & dosage , Melanoma/secondary , Melanoma/therapy , Radiosurgery/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/adverse effects , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Female , Humans , Immune Tolerance , Ipilimumab/adverse effects , Male , Maximum Tolerated Dose , Melanoma/immunology , Middle Aged , Radiotherapy Dosage , Response Evaluation Criteria in Solid Tumors , Young Adult
3.
Exp Dermatol ; 27(5): 463-469, 2018 05.
Article in English | MEDLINE | ID: mdl-28833576

ABSTRACT

RNA interference has emerged as a powerful tool for therapeutic gene silencing, as it offers the possibility to silence virtually any known pathology-causing gene. However, in vivo delivery of RNAi molecules is hampered by their unfavourable physicochemical characteristics and susceptibility to degradation by endogenous enzymes. To overcome these limitations, we recently developed an elastic liposomal formulation, called DDC642, as topical delivery system of therapeutic RNAi molecules for skin disorders. In this study, we validated the therapeutic efficacy of DDC642-encapsulated RNAi molecules in the treatment of psoriasis using 3 different in vitro models: a standardized keratinocyte monolayer culture, psoriasis-induced keratinocytes and a psoriasis-reconstructed skin model. Four genes (IL22RA1, KRT17, DEFB4 and TSLP), known to be upregulated in psoriatic lesions, and thereby key players in psoriasis pathogenesis were selected. Moreover, the possibility of using a combined siRNA therapy in the topical treatment of psoriasis was explored. Results indicate a successful gene silencing of each different target, both at mRNA and protein levels. Additionally, siRNA-DDC642 treatment resulted in a reduced expression of specific psoriasis markers, indicating their potential in future therapeutic approach. The examined siRNA combination (ie simultaneous knockdown of KRT17, DEFB4 and TSLP) showed an enhanced reduction in TSLP expression, whereas the decrease in K17 protein expression was impaired in psoriatic keratinocytes. Although the here examined siRNA combination could still be further improved, our study proved already in vitro the clinical potential of targeting multiple genes at once, each playing a different role in a complex disease such as psoriasis.


Subject(s)
In Vitro Techniques , Models, Biological , Psoriasis/therapy , RNAi Therapeutics , Adult , Biomarkers/metabolism , Humans , Keratinocytes/metabolism , Proof of Concept Study
4.
J Transl Med ; 15(1): 150, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28662677

ABSTRACT

BACKGROUND: Current first-line standard of therapy for metastatic urothelial carcinoma is platinum-based combination chemotherapy. Pembrolizumab in phase III has demonstrated a promising overall response rate of 21.1% in patients with progression or recurrence after platinum-based chemotherapy. Preclinical and clinical evidence suggests that radiotherapy has a systemic anti-cancer immune effect and can increase the level of PD-L1 and tumor infiltrating lymphocytes in the tumor microenvironment. These findings gave rise to the hypothesis that the combination of radiotherapy with anti-PD1 treatment could lead to a synergistic effect, hereby enhancing response rates. METHODS: The phase I part will assess the dose limiting toxicity of the combination treatment of stereotactic body radiotherapy (SBRT) with four cycles of pembrolizumab (200 mg intravenously, every 3 weeks) in patients with metastatic urothelial carcinoma. The dose of both pembrolizumab and SBRT will be fixed, yet the patients will be randomized to receive SBRT either before the first cycle of pembrolizumab or before the third cycle of pembrolizumab. SBRT will be delivered (24 Gy in 3 fractions every other day) to the largest metastatic lesion. Secondary objectives include response rate according to RECIST v1.1 and immune related response criteria, progression-free survival and overall survival. The systemic immune effect triggered by the combination therapy will be monitored on various time points during the trial. The PD-L1/TIL status of the tumors will be analyzed via immunohistochemistry and response rates in the subgroups will be analyzed separately. A Simon's two-stage optimum design is used to select the treatment arm associated with the best response rate and with acceptable toxicity to proceed to the phase II trial. In this phase, 13 additional patients will be accrued to receive study treatment. DISCUSSION: The progress made in the field of immunotherapy has lead to promising breakthroughs in various solid malignancies. Unfortunately, the majority of patients do not respond. The current trial will shed light on the toxicity and potential anti-tumor activity of the combination of radiotherapy with anti-PD1 treatment and may identify potential new markers for response and resistance to therapy. Trial registration this trial is registered on clinicaltrials.gov (NCT02826564).


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Radiosurgery/adverse effects , Urologic Neoplasms/immunology , Urologic Neoplasms/therapy , Urothelium/pathology , Combined Modality Therapy , Dose-Response Relationship, Radiation , Female , Follow-Up Studies , Humans , Male , Neoplasm Metastasis , Sample Size , Statistics as Topic , Urologic Neoplasms/drug therapy , Urologic Neoplasms/radiotherapy
5.
Exp Biol Med (Maywood) ; 242(11): 1158-1169, 2017 06.
Article in English | MEDLINE | ID: mdl-28585891

ABSTRACT

Psoriasis is a complex chronic immune-mediated inflammatory cutaneous disease associated with the development of inflammatory plaques on the skin. Studies proved that the disease results from a deregulated interplay between skin keratinocytes, immune cells and the environment leading to a persisting inflammatory process modulated by pro-inflammatory cytokines and activation of T cells. However, a major hindrance to study the pathogenesis of psoriasis more in depth and subsequent development of novel therapies is the lack of suitable pre-clinical models mimicking the complex phenotype of this skin disorder. Recent advances in and optimization of three-dimensional skin equivalent models have made them attractive and promising alternatives to the simplistic monolayer cultures, immunological different in vivo models and scarce ex vivo skin explants. Moreover, human skin equivalents are increasing in complexity level to match human biology as closely as possible. Here, we critically review the different types of three-dimensional skin models of psoriasis with relevance to their application potential and advantages over other models. This will guide researchers in choosing the most suitable psoriasis skin model for therapeutic drug testing (including gene therapy via siRNA molecules), or to examine biological features contributing to the pathology of psoriasis. However, the addition of T cells (as recently applied to a de-epidermized dermis-based psoriatic skin model) or other immune cells would make them even more attractive models and broaden their application potential. Eventually, the ultimate goal would be to substitute animal models by three-dimensional psoriatic skin models in the pre-clinical phases of anti-psoriasis candidate drugs. Impact statement The continuous development of novel in vitro models mimicking the psoriasis phenotype is important in the field of psoriasis research, as currently no model exists that completely matches the in vivo psoriasis skin or the disease pathology. This work provides a complete overview of the different available in vitro psoriasis models and suggests improvements for future models. Moreover, a focus was given to psoriatic skin equivalent models, as they offer several advantages over the other models, including commercial availability and validity. The potential and reported applicability of these models in psoriasis pre-clinical research is extensively discussed. As such, this work offers a guide to researchers in their choice of pre-clinical psoriasis model depending on their type of research question.


Subject(s)
Models, Biological , Organ Culture Techniques/methods , Psoriasis/physiopathology , Skin/pathology , Tissue Engineering/methods , Humans
6.
J Transl Med ; 15(1): 21, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137295

ABSTRACT

BACKGROUND: Antibodies blocking programmed cell death 1 (PD-1) have encouraging responses in patients with metastatic melanoma. Response to anti-PD-1 treatment requires pre-existing CD8+ T cells that are negatively regulated by PD-1-mediated adaptive immune resistance. Unfortunately, less than half of melanoma tumours have these characteristics. Combining anti-PD-1 treatment with other immunomodulating treatments to activate CD8+ T cells is therefore of vital importance to increase response rates and long-term survival benefit in melanoma patients. Both preclinical and retrospective clinical data support the hypothesis that radiotherapy increases the response rates to anti-PD-1 treatment by stimulating the accumulation and activation of CD8+ T cells in the tumour microenvironment. Combining radiotherapy with a PD-1 blocking antibody might therefore increase response rates and even induce long-term survival. The current phase II study will be testing these hypotheses and aims to improve local and distant tumour responses by exploiting the pro-immunogenic effects of radiotherapy in addition to anti-PD-1 treatment. METHODS: The trial will be conducted in patients with metastatic melanoma. Nivolumab or pembrolizumab, both antibodies that target PD-1, will be administrated according to the recommended dosing schedule. Prior to the 2nd cycle, radiotherapy will be delivered in three fractions of 8 Gy to the largest FDG-avid metastatic lesion. The primary endpoint is the proportion of patients with a partial or complete response in non-irradiated metastases according to RECIST v1.1. Secondary endpoints include response rate according to immune related response criteria, metabolic response, local control and survival. To identify peripheral blood biomarkers, peripheral blood mononuclear cells and serum samples will be collected prospectively before, during and after treatment and subjected to flow cytometry and cytokine measurement. DISCUSSION: The current phase II trial aims at exploring the suggested benefits of combining anti-PD-1 treatment and radiotherapy. The translational focus on immunologic markers might be suitable for predicting efficacy and monitoring the effect so to improve patient selection for future clinical applications. ClinicalTrials.gov Identifier NCT02821182.


Subject(s)
Clinical Trials, Phase II as Topic , Melanoma/secondary , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Radiosurgery , Endpoint Determination , Follow-Up Studies , Humans , Melanoma/immunology , Outcome Assessment, Health Care , Programmed Cell Death 1 Receptor/metabolism , Sample Size , Treatment Outcome
7.
Expert Opin Drug Deliv ; 14(1): 109-122, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27348356

ABSTRACT

INTRODUCTION: In the treatment of dermatological disorders, topical drug administration is a mainstay. However, nanoparticle-based carrier systems could improve and expand the current therapeutic range via localized delivery of active ingredients. Areas covered: This review gives a detailed description of lipid- and surfactant-based drug delivery systems which have been explored for topical drug administration. To guide researchers in their choice of delivery system, an informative decision tree is included. Moreover, this review provides a complete overview of the topical or transdermal drug products, currently on the market or under clinical investigation, delivered via the discussed carriers, in the treatment of skin disorders. Expert opinion: Conventional liposomes are still popular in the domain of topical or transdermal drug delivery and dominate the market landscape. However, several other carriers, such as exosomes and niosomes, are being explored which offer distinct advantages over liposomes and should therefore not be disregarded when selecting a proper drug delivery system.


Subject(s)
Drug Delivery Systems , Nanoparticles , Skin Diseases/drug therapy , Administration, Cutaneous , Humans , Lipids/chemistry , Liposomes/metabolism , Skin/metabolism , Skin Absorption , Surface-Active Agents/chemistry
8.
PLoS One ; 11(10): e0164080, 2016.
Article in English | MEDLINE | ID: mdl-27711196

ABSTRACT

The recent interest and elucidation of the JAK/STAT signaling pathway created new targets for the treatment of inflammatory skin diseases (ISDs). JAK inhibitors in oral and topical formulations have shown beneficial results in psoriasis and alopecia areata. Patients suffering from other ISDs might also benefit from JAK inhibition. Given the development of specific JAK inhibitors, the expression patterns of JAKs in different ISDs needs to be clarified. We aimed to analyze the expression of JAK/STAT family members in a set of prevalent ISDs: psoriasis, lichen planus (LP), cutaneous lupus erythematosus (CLE), atopic dermatitis (AD), pyoderma gangrenosum (PG) and alopecia areata (AA) versus healthy controls for (p)JAK1, (p)JAK2, (p)JAK3, (p)TYK2, pSTAT1, pSTAT2 and pSTAT3. The epidermis carried in all ISDs, except for CLE, a strong JAK3 signature. The dermal infiltrate showed a more diverse expression pattern. JAK1, JAK2 and JAK3 were significantly overexpressed in PG and AD suggesting the need for pan-JAK inhibitors. In contrast, psoriasis and LP showed only JAK1 and JAK3 upregulation, while AA and CLE were characterized by a single dermal JAK signal (pJAK3 and pJAK1, respectively). This indicates that the latter diseases may benefit from more targeted JAK inhibitors. Our in vitro keratinocyte psoriasis model displayed reversal of the psoriatic JAK profile following tofacitinib treatment. This direct interaction with keratinocytes may decrease the need for deep skin penetration of topical JAK inhibitors in order to exert its effects on dermal immune cells. In conclusion, these results point to the important contribution of the JAK/STAT pathway in several ISDs. Considering the epidermal JAK3 expression levels, great interest should go to the investigation of topical JAK3 inhibitors as therapeutic option of ISDs.


Subject(s)
Janus Kinase 3/metabolism , Molecular Targeted Therapy , Skin Diseases/drug therapy , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Infant , Inflammation/complications , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Diseases/complications , Skin Diseases/enzymology , Young Adult
9.
Data Brief ; 7: 1204-10, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27408923

ABSTRACT

This article contains original data, figures and methods used in the characterization of the liposomal carrier 'DDC642' for topical applications, described in "An elastic liposomal formulation for RNAi-based topical treatment of skin disorders: proof-of-concept in the treatment of psoriasis" (Desmet et al., 2016) [1]. Several elastic liposomal formulations have been evaluated for their ability to encapsulate and deliver RNA interference (RNAi) molecules to cultured primary skin cells. The efficiency and effectiveness of these liposomes were compared to that of our previously characterized liposomes, the 'SECosomes' (SEC) (Geusens et al., 2010) [2]. After selection of a potential superior carrier, based on encapsulation and transfection efficiency data (Desmet et al., 2016) [1], the selected DDC642 liposomes were characterized more in-depth. Herein, a detailed characterization of the DDC642 liposome and RNAi-loaded lipoplexes is given, including the matching protocols.

11.
Int J Pharm ; 500(1-2): 268-74, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26806466

ABSTRACT

RNA interference (RNAi) is a rapidly emerging approach for targeted gene silencing to alleviate disease pathology. However, lack of efficient carriers for targeted delivery delays the clinical translation of RNAi. An interesting target for local RNAi therapeutics is the skin as it allows direct access to target cells. Still, applications are limited due to the effective skin barrier which hinders penetration. Herein, a description is given of a liposomal carrier, called 'DDC642', capable of delivering RNAi molecules to the epidermis of impaired and intact human skin, without targeting the dermis or circulatory system. In a psoriasis tissue model, down-regulation of the psoriasis marker human beta-defensin 2 by DDC642-delivered siRNA was confirmed, providing proof-of-concept. These liposomes thus hold great potential as topical delivery system for RNAi therapeutics in the treatment of numerous skin diseases.


Subject(s)
Gene Transfer Techniques , Psoriasis/therapy , RNA Interference , Administration, Topical , Cholesterol/chemistry , Elasticity , Ethanol/chemistry , Fatty Acids, Monounsaturated/chemistry , Humans , Keratinocytes/metabolism , Liposomes , Melanocytes/metabolism , MicroRNAs/administration & dosage , Phosphatidylethanolamines/chemistry , Psoriasis/genetics , Quaternary Ammonium Compounds/chemistry , RNA, Small Interfering/administration & dosage , beta-Defensins/genetics
12.
Melanoma Res ; 25(4): 357-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050145

ABSTRACT

Interferon-α (IFN-α) is the only approved adjuvant treatment for high-risk melanoma patients in Europe, but the impact on overall survival is low. Although it is believed that IFN-α exerts its effects through immunomodulation, data on its impact on circulating immune cells are scarce. Flow cytometry was performed on peripheral blood mononuclear cells of eight IFN-α2b-treated stage III melanoma patients and 26 untreated stage III melanoma patients as controls to enumerate myeloid and plasmacytoid dendritic cells (mDC and pDC), monocytic and polymorphonuclear myeloid-derived suppressor cells (mMDSC and pmnMDSC) and cytotoxic and regulatory T-cells (Tregs). The expression of several immunosuppressive markers [indoleamine 2,3-dioxygenase (IDO), programmed-death ligand-1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA4)] was explored. IDO activity in the blood was confirmed by ultra-performance liquid chromatography. Compared with controls, IFN-α2b treatment was associated with increased IDO expression by pDCs (P=0.021) and an increased kynurenine/tryptophan ratio in the serum (P=0.004), compatible with IDO enzyme activity. Furthermore, IFN-α2b-treated patients had a decreased mDC/DC ratio (P=0.002), decreased CD3+ lymphocytes (P=0.034) and increased circulating Treg (P<0.001) and PD-L1+cytotoxic T-cell (P=0.001) frequencies. IDO expression is upregulated in circulating pDCs of high-risk melanoma patients treated with adjuvant IFN-α2b. This is associated with tryptophan consumption in the patients' serum and higher Treg and PD-L1+cytotoxic T-cell frequencies. We hypothesize that in IFN-α2b-treated patients, IDO activity acts as a negative feedback mechanism and might limit the clinical efficacy of IFN-α2b therapy. The underlying mechanism should be explored as this could lead to more efficient immunotherapies.


Subject(s)
Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-alpha/therapeutic use , Melanoma/drug therapy , Myeloid Cells/immunology , Adult , Aged , B7-H1 Antigen/biosynthesis , CTLA-4 Antigen/biosynthesis , Europe , Female , Flow Cytometry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Leukocytes, Mononuclear/immunology , Male , Melanoma/immunology , Middle Aged , Neoplasm Staging , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Melanoma, Cutaneous Malignant
13.
J Transl Med ; 13: 9, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25592374

ABSTRACT

BACKGROUND: Immune markers in the peripheral blood of melanoma patients could provide prognostic information. However, there is currently no consensus on which circulating cell types have more clinical impact. We therefore evaluated myeloid-derived suppressor cells (MDSC), dendritic cells (DC), cytotoxic T-cells and regulatory T-cells (Treg) in a series of blood samples of melanoma patients in different stages of disease. METHODS: Flow cytometry was performed on peripheral blood mononuclear cells of 69 stage I to IV melanoma patients with a median follow-up of 39 months after diagnosis to measure the percentage of monocytic MDSCs (mMDSCs), polymorphonuclear MDSCs (pmnMDSCs), myeloid DCs (mDCs), plasmacytoid DCs (pDCs), cytotoxic T-cells and Tregs. We also assessed the expression of PD-L1 and CTLA-4 in cytotoxic T-cells and Tregs respectively. The impact of cell frequencies on prognosis was tested with multivariate Cox regression modelling. RESULTS: Circulating pDC levels were decreased in patients with advanced (P = 0.001) or active (P = 0.002) disease. Low pDC levels conferred an independent negative impact on overall (P = 0.025) and progression-free survival (P = 0.036). Even before relapse, a decrease in pDC levels was observed (P = 0.002, correlation coefficient 0.898). High levels of circulating MDSCs (>4.13%) have an independent negative prognostic impact on OS (P = 0.012). MDSC levels were associated with decreased CD3+ (P < 0.001) and CD3 + CD8+ (P = 0.017) T-cell levels. Conversely, patients with high MDSC levels had more PD-L1+ T-cells (P = 0.033) and more CTLA-4 expression by Tregs (P = 0.003). pDCs and MDSCs were inversely correlated (P = 0.004). The impact of pDC levels on prognosis and prediction of the presence of systemic disease was stronger than that of MDSC levels. CONCLUSION: We demonstrated that circulating pDC and MDSC levels are inversely correlated but have an independent prognostic value in melanoma patients. These cell types represent a single immunologic system and should be evaluated together. Both are key players in the immunological climate in melanoma patients, as they are correlated with circulating cytotoxic and regulatory T-cells. Circulating pDC and MDSC levels should be considered in future immunoprofiling efforts as they could impact disease management.


Subject(s)
Dendritic Cells/immunology , Melanoma/immunology , Myeloid Cells/immunology , Adult , Cell Movement , Disease-Free Survival , Humans , Logistic Models , Melanoma/pathology , Middle Aged , Neoplasm Staging , Proportional Hazards Models , Skin Neoplasms , Melanoma, Cutaneous Malignant
14.
Appl Microbiol Biotechnol ; 98(15): 6831-46, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24917376

ABSTRACT

Crosstalk between the human host and its microbiota is reported to influence various diseases such as mucositis. Fundamental research in this area is however complicated by the time frame restrictions during which host-microbe interactions can be studied in vitro. The model proposed in this paper, consisting of an oral epithelium and biofilm, can be used to study microbe-host crosstalk in vitro in non-infectious conditions up to 72 h. Microbiota derived from oral swabs were cultured on an agar/mucin layer and challenged with monolayers of keratinocytes grown on plastic or collagen type I layers embedded with fibroblasts. The overall microbial biofilm composition in terms of diversity remained representative for the oral microbiome, whilst the epithelial cell morphology and viability were unaffected. Applying the model to investigate wound healing revealed a reduced healing of 30 % in the presence of microbiota, which was not caused by a reduction of the proliferation index (52.1-61.5) or a significantly increased number of apoptotic (1-1.13) or necrotic (32-30.5 %) cells. Since the model allows the separate study of the microbial and cellular exometabolome, the biofilm and epithelial characteristics after co-culturing, it is applicable for investigations within fundamental research and for the discovery and development of agents that promote wound healing.


Subject(s)
Microbiota , Mouth Diseases/physiopathology , Mouth Mucosa/microbiology , Wound Healing , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Biofilms , Cell Line , Cell Proliferation , Cells, Cultured , Host-Pathogen Interactions , Humans , Keratinocytes/microbiology , Mice , Models, Biological , Mouth Diseases/microbiology , Mouth Mucosa/physiology
15.
Pigment Cell Melanoma Res ; 27(4): 512-24, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24612852

ABSTRACT

Hyperpigmentation is a key feature in a variety of inherited and acquired syndromes. Nonetheless, determining the exact diagnosis only on the clinical phenotype can be challenging, and a detailed search for associated symptoms is often of crucial importance. As pigmentation pathways are regulated by complex signaling transduction cascades (e.g. MSH/cAMP, KIT signaling pathways), the underlying defects leading to elevated melanin production are numerous. With regard to treatment, limited therapeutic options exist, each with specific side effects. In acquired hyperpigmentation, the melanin deposition may, however, be reversible after adequate therapy of the underlying disorder or even disappear spontaneously. In this review, we provide an overview of the biology of hyperpigmentation syndromes classified according to the main underlying defect that deregulates physiological melanogenesis. The identification of novel genes or key players involved in hyperpigmentary disorders is becoming increasingly important in view of the development of safer and more efficient treatments.


Subject(s)
Genetic Diseases, Inborn/metabolism , Hyperpigmentation/metabolism , Melanins/metabolism , Signal Transduction , Animals , Genetic Diseases, Inborn/classification , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Humans , Hyperpigmentation/classification , Hyperpigmentation/diagnosis , Hyperpigmentation/genetics , Melanins/genetics , Syndrome
16.
Oncoscience ; 1(10): 634-48, 2014.
Article in English | MEDLINE | ID: mdl-25593993

ABSTRACT

Cancer progression is characterized by a complex reciprocity between neoplastic epithelium and adjacent stromal cells. In ductal carcinoma in situ (DCIS) of the breast, both reduced stromal decorin expression and myxoid stroma are correlated with increased recurrence risk. In this study, we aimed to investigate paracrine regulation of expression of decorin and related extracellular matrix (ECM) proteins in cancer-associated fibroblasts (CAFs). Transforming growth factor-ß1 (TGF-ß1) was identified as a competent ECM modulator, as it reduced decorin and strongly enhanced versican, biglycan and type I collagen expression. Similar but less pronounced effects were observed when fibroblasts were treated with basic fibroblast growth factor (bFGF). Despite this concerted ECM modulation, TGF-ß1 and bFGF differentially regulated alpha-smooth muscle actin (α-SMA) expression, which is often proposed as a CAF-marker. Cancer cell-derived secretomes induced versican and biglycan expression in fibroblasts. Immunohistochemistry on twenty DCIS specimens showed a trend toward periductal versican overexpression in DCIS with myxoid stroma. Cancer cell adhesion was inhibited by decorin, but not by CAF-derived matrices. Cancer cells presented significantly enhanced spreading when seeded on matrices derived from TGF-ß1-treated CAF. Altogether these data indicate that preinvasive cancerous lesions might modulate the composition of surrounding stroma through TGF-ß1 release to obtain an invasion-permissive microenvironment.

17.
PLoS One ; 8(8): e70538, 2013.
Article in English | MEDLINE | ID: mdl-23950955

ABSTRACT

The skin microbial community is regarded as essential for human health and well-being, but likewise plays an important role in the formation of body odor in, for instance, the axillae. Few molecular-based research was done on the axillary microbiome. This study typified the axillary microbiome of a group of 53 healthy subjects. A profound view was obtained of the interpersonal, intrapersonal and temporal diversity of the human axillary microbiota. Denaturing gradient gel electrophoresis (DGGE) and next generation sequencing on 16S rRNA gene region were combined and used as extent to each other. Two important clusters were characterized, where Staphylococcus and Corynebacterium species were the abundant species. Females predominantly clustered within the Staphylococcus cluster (87%, n = 17), whereas males clustered more in the Corynebacterium cluster (39%, n = 36). The axillary microbiota was unique to each individual. Left-right asymmetry occurred in about half of the human population. For the first time, an elaborate study was performed on the dynamics of the axillary microbiome. A relatively stable axillary microbiome was noticed, although a few subjects evolved towards another stable community. The deodorant usage had a proportional linear influence on the species diversity of the axillary microbiome.


Subject(s)
Axilla/microbiology , Corynebacterium/classification , Staphylococcus/classification , Biodiversity , Cluster Analysis , Corynebacterium/genetics , Denaturing Gradient Gel Electrophoresis , Female , Humans , Male , Microbiota , Phylogeny , Skin/microbiology , Staphylococcus/genetics
18.
Int J Oncol ; 42(4): 1443-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404256

ABSTRACT

MicroRNAs (miRNAs) are post-transcriptional modulators of gene expression which play important roles in tumorigenesis and cancer metastasis. Since they are often highly deregulated in various types of cancer, miRNAs may be effective treatment targets. miRNA profiling studies of melanoma have led to the identification of several tumor suppressor miRNAs. One of these include miR-145, although functional data proving its specific function are limited. Therefore, in this study, we examined the expression levels of miR-145 in three melanoma cell lines (BLM, FM3P and WM793). Additional gain-of-function experiments revealed that miR-145 exerts an anti-proliferative effect in the primary, non-invasive melanoma cell line, WM793, whereas cell migration and the invasive potential of metastatic melanoma cells was suppressed following transfection with miR-145 mimics. In order to investigate the mechanisms by which miR-145 exerts its invasion suppressor function, we examined the expression level of target genes [fascin homolog 1 (FSCN1), myosin­Va (MYO5A and SOX9] and that of an indirect target (RAB27A) following the overexpression of miR-145. The results showed that SOX9, MYO5A and RAB27A were not involved in the biological effects caused by miR-145 mimics. Surprisingly, we discovered that miR-145 in melanoma, in contrast to many other tumor types, does not necessarily act via the target, FSCN1, since the downregulation of FSCN1 did not inhibit cell proliferation or migration but, on the contrary, increased cell invasion in two out of the three melanoma cell lines examined. Our in vitro data is in accordance with previously reported in vivo data describing the low expression of FSCN1 in malignant melanomas when compared to dysplastic nevi, suggesting that the expression of FSCN1 decreases as the formation and progression stage of melanoma advances. In conclusion, our data provide evidence that miR-145 is an invasion suppressor in metastatic melanoma cells. Despite the fact that it remains unclear which genes or pathways are regulated by miR-145 in melanoma, miR-145 may serve as a useful therapeutic agent in melanoma when re-expressed in situ.


Subject(s)
Cell Movement , Melanoma/secondary , MicroRNAs/metabolism , Skin Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Melanocytes/metabolism , Melanoma/metabolism , MicroRNAs/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasm Invasiveness , RNA Interference , Skin Neoplasms/metabolism
19.
J Invest Dermatol ; 133(1): 201-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22895360

ABSTRACT

The current treatments for hyperpigmentation are often associated with a lack of efficacy and adverse side effects. We hypothesized that microRNA (miRNA)-based treatments may offer an attractive alternative by specifically targeting key genes in melanogenesis. The aim of this study was to identify miRNAs interfering with the pigmentary process and to assess their functional role. miRNA profiling was performed on mouse melanocytes after three consecutive treatments involving forskolin and solar-simulated UV (ssUV) irradiation. Sixteen miRNAs were identified as differentially expressed in treated melan-a cells versus untreated cells. Remarkably, a 15-fold downregulation of miR-145 was detected. Overexpression or downregulation of miR-145 in melan-a cells revealed reduced or increased expression of Sox9, Mitf, Tyr, Trp1, Myo5a, Rab27a, and Fscn1, respectively. Moreover, a luciferase reporter assay demonstrated direct targeting of Myo5a by miR-145 in mouse and human melanocytes. Immunofluorescence tagging of melanosomes in miR-145-transfected human melanocytes displayed perinuclear accumulation of melanosomes with additional hypopigmentation of harvested cell pellets. In conclusion, this study has established an miRNA signature associated with forskolin and ssUV treatment. The significant down- or upregulation of major pigmentation genes, after modulating miR-145 expression, suggests a key role for miR-145 in regulating melanogenesis.


Subject(s)
MicroRNAs/biosynthesis , Pigmentation/physiology , Animals , Cells, Cultured , Colforsin/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , MART-1 Antigen/analysis , MART-1 Antigen/genetics , MART-1 Antigen/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanosomes/genetics , Melanosomes/metabolism , Mice , MicroRNAs/genetics , Microphthalmia-Associated Transcription Factor/biosynthesis , Monophenol Monooxygenase/biosynthesis , Myosin Heavy Chains/biosynthesis , Myosin Type V/biosynthesis , Pigmentation/genetics , SOX9 Transcription Factor/biosynthesis , Transfection , Trypsin/biosynthesis , Ultraviolet Rays , rab GTP-Binding Proteins/biosynthesis , rab27 GTP-Binding Proteins
20.
Oncoimmunology ; 1(4): 526-528, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22754773

ABSTRACT

Insights into immune reactions against benign and malignant melanocytes may help the development of more efficient immunotherapeutic treatments for melanoma. The interplay between an active systemic antitumor immunity and a responsive local tumor environment is crucial to achieve effective clinical responses. Increasing evidence confirms this strategy can lead to an adequate and durable immunosurveillance of melanocytes.

SELECTION OF CITATIONS
SEARCH DETAIL
...