Hans-Jürgen Stark’s research while affiliated with Heidelberg University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (64)


miRNA-29-CLIP uncovers new targets and functions to improve skin repair
  • Preprint

May 2024

·

30 Reads

Lalitha Thiagarajan

·

Rosa Sanchez-Alvarez

·

Chiho Kambara

·

[...]

·

MicroRNAs (miRNAs) control organogenesis in mammals but their role in specific cell types is not fully explored. miRNAs exert their function by binding mRNAs and inhibiting translation. Skin is an excellent model to study the role of miRNAs in primary cells of epidermal (keratinocytes) and mesodermal (fibroblasts) origin, because the growth of these cells is tightly controlled at translation. Previous research demonstrated that miRNA-29 family functions during skin repair, however, the exact mRNA targets and the downstream mechanisms of miRNA-29-mediated regulation of cell growth is missing. Here, we use miRNA crosslinking and immunoprecipitation (miRNA-CLIP) method to find the direct targets of miRNA-29 in keratinocytes and fibroblasts from human skin. We uncover previously unrecognized roles of miRNA-29 in protein folding and RNA processing, common to all cell types tested, and determine the cell-specific role of miRNA-29. Using modified anti-sense oligonucleotides (ASO) in 2D and 3D cultures of keratinocytes and fibroblasts, we enhanced cell-to-matrix adhesion and found an autocrine and paracrine mechanism of miRNA-29-dependent cell growth. Our results include a comprehensive list and functional analyses of mRNAs directly bound by miRNA-29 keratinocytes and fibroblasts, determined by miRNA-CLIP and ASO-mediated inhibition of miRNA-29 followed by RNA-seq. We reveal a full transcriptome of human keratinocytes with enhanced adhesion to the wound matrix, which supports regeneration of the epidermis and is regulated by miRNA-29. The functions of miRNA-29 identified in this study can provide a new approach to improve cutaneous repair by restoring and enhancing the endogenous mechanisms through the stage-specific delivery of miRNA-29 ASO.


Vemurafenib-dependent pathway regulation and proliferation. A375 (BRAF mutant) melanoma cells (A) and BRAF wildtype keratinocytes, represented by normal human keratinocytes (NHEK) (B), HaCaT (C), the premalignant HrasA5 (D), and the malignant HrasII4 cells (E), were incubated with either DMSO (solvent control) or vemurafenib (1 µM) for a period of up to 6 h, and protein expression of MEK/P-MEK, ERK/P-ERK, p38/P-p38, and Akt/PAkt was examined. Opposing effects were seen for P-MEK and P-ERK when comparing the A375 melanoma cells (BRAF-V600E mut) and the human keratinocytes. P-p38 and P-Akt were inhibited transiently in the A375 cells, both in control and vemurafenib-treated cells, but did not seem to be regulated in the human keratinocytes. GAPDH was used as loading control in all immunoblots. To study the effects on proliferation, NHEK (F), HaCaT (G), and HrasA5 cells (H) were treated with different concentrations of vemurafenib (0.01–5 μM) for a period of up to 72 h, and proliferation was determined at 24, 48, and 72 h by measuring fluorescence intensity (SyBr green proliferation assay). Statistical significance was calculated by two-way ANOVA and Bonferroni posttest (n = 2, mean ± SD, two-way ANOVA + Bonferroni posttest; ns, not significant).
Vemurafenib preferentially targets the degradome. Vemurafenib-altered expression profile of NHEK (A), HaCaT (B), and HrasA5 cells (C) after 8 and 24 h of vemurafenib treatment. All human keratinocytes show upregulation of IL1A and IL1B, with HaCaT cells being the least regulated, as well as a strong induction of MMP1 and MMP3 while MMP9 and MMP14 remain largely unaffected. In addition, epidermal differentiation markers become upregulated in NHEK only. Normalization was performed using GAPDH as house-keeping gene and foldchanges were expressed by comparing 1 or 5 µM vemurafenib treatment of NHDF to DMSO stimulation, respectively. n = 2, mean ± SD.
Characterization of the effects of vemurafenib on epithelial differentiation and proliferation in SEs. SEs from NHEK and HrasA5 cells were treated with vemurafenib (VEM) (1 and 5 µM) for up to 5 weeks and histology and immunostaining were performed at the indicated time points. (A) H&E staining of SEs from NHEK demonstrates accelerated cornification, particularly evident upon 5 µM VEM (left). Also, HrasA5 epithelia showed a time-dependent increase in cornification. In addition, invasion was seen after 3 (1 µM VEM) and 1 week (5 µM VEM), respectively (right). (B) Improved differentiation is confirmed by immunostaining for the early, KRT10, and the late differentiation markers FLG and KRT2 in the epithelium of the NHEKs (left) and the HrasA5 cells (right). The BM components COLVII (green), COLIV (red), and LAM (red) are expressed as contiguous lines in NHEK SEs at all time points and all conditions. Note that COLIV is expressed continuously and present throughout the DE; though enriched in the BM zone (left). In HrasA5 SEs, COLVII is generally reduced and lost at the invasive front. COLIV and LAM rather appear “bloated” with the tumor cells pushing through small gaps (right). (C) Same SEs stained for the proliferation marker H3S10ph (red), demonstrating very similar proliferation for all NHEK SEs (left). In HrasA5 SEs, proliferation is present throughout the epithelium (control). Under VEM, proliferation gets restricted to the basal compartment (right). All SEs are counterstained for the early differentiation marker transglutaminase 1 (TGM). (D) For quantification of proliferation SEs from NHEK and HrasA5 cells were costained with COLVII and KI67 and the number of proliferating cells (KI67+) correlated with BM (COLVII) length. Neither NHEKs nor HrasA5 cells showed a significant regulation in proliferation (n = 2, mean + SEM, one-way ANOVA + Dunnett’s multiple comparison test; ns, not significant). For all conditions, nuclei were counterstained with DAPI (blue). Treatment with the DMSO was used as control. Time specifications relate to time after starting the scale bar = 300 µm [for (A, C)]; scale bar = 150 µm [for (B)].
Vemurafenib-dependent degradome. SEs from NHEK, HaCaT cells, and HrasA5 cells were treated with vemurafenib (VEM) (5 µM) for 3 weeks and sections of control (DMSO) and VEM-treated SEs were stained for MMP1 and vimentin (A) and MMP3 (B). Nuclei were counterstained with DAPI. All VEM-treated samples show a clear upregulation of both MMPs scale bar = 150 µm. (C) ELISA for HaCaT and HrasA5 SEs treated with DMSO (control), cobimetinib (COBI), VEM, and the combination of COBI and VEM with the indicated concentrations. Secretion of MMP1, MMP3, and MMP9 were quantified (n = 3, mean ± SD, one-way ANOVA + Dunnett’s multiple comparison test; ns, not significant). (D) Gelatinase assay confirms a strong proteolytic activity (green) in the VEM-treated (5 µM) HrasA5 SEs compared with the untreated controls. Scale bar = 300 μm.
Inhibition of the vemurafenib-dependent degradome in HrasA5 SEs by suppressing MMP activity. (A) H&E-stained HrasA5-SEs under vemurafenib (5 µM) exhibit an invasive phenotype with signs of subepithelial disintegration under the invading pegs (left, arrow), a physiological state not seen in SEs treated in addition with the MMP inhibitor ilomastat (10 µM) (right). (B) Staining for the BM-components LAM (red, above) and ColVII (green, bottom) points to a pronounced degradation upon vemurafenib (middle) when compared with untreated control SEs (left). Additional application of ilomastat prevents proteolysis and allows for an uninterrupted BM (right). Likewise, delayed onset of KRT10 expression (green, middle, above) is widely renormalized by ilomastat (right, above). (C) Beyond the BM, also the subepithelial extracellular matrix is affected by the increased proteolytic activity. Picrosirius red staining visualizes semiquantitatively the amount of stromal collagen in bright field microscopy (above) and even more clearly in circular polarization microscopy that specifically highlights organized collagen bundles (bottom). Whereas under vemurafenib the density of collagen fibers is drastically reduced to 48.80% (middle), cotreatment with ilomastat completely preserves the control state (100.85% vs. 100%, right vs. left). Scale bar = 300 µM.

+3

Adverse Effects of Vemurafenib on Skin Integrity: Hyperkeratosis and Skin Cancer Initiation Due to Altered MEK/ERK-Signaling and MMP Activity
  • Article
  • Full-text available

January 2022

·

143 Reads

·

4 Citations

The BRAF inhibitor vemurafenib, approved for treating patients with BRAF V600E-mutant and unresectable or metastatic melanomas, rapidly induces cutaneous adverse events, including hyperkeratotic skin lesions and cutaneous squamous cell carcinomas (cSCC). To determine, how vemurafenib would provoke these adverse events, we utilized long-term in vitro skin equivalents (SEs) comprising epidermal keratinocytes and dermal fibroblasts in their physiological environment. We inserted keratinocytes with different genetic background [normal keratinocytes: NHEK, HaCaT (p53/mut), and HrasA5 (p53/mut+Hras/mut)] to analyze effects depending on the stage of carcinogenesis. We now show that vemurafenib activates MEK-ERK signaling in both, keratinocytes, and fibroblasts in vitro and in the in vivo-like SEs. As a consequence, vemurafenib does not provide a growth advantage but leads to a differentiation phenotype, causing accelerated differentiation and hyperkeratosis in the NHEK and normalized stratification and cornification in the transformed keratinocytes. Although all keratinocytes responded very similarly to vemurafenib in their expression profile, particularly with a significant induction of MMP1 and MMP3, only the HrasA5 cells revealed a vemurafenib-dependent pathophysiological shift to tumor progression, i.e., the initiation of invasive growth. This was shown by increased proteolytic activity allowing for penetration of the basement membrane and invasion into the disrupted underlying matrix. Blocking MMP activity, by the addition of ilomastat, prevented invasion with all corresponding degradative activities, thus substantiating that the RAS-RAF-MEK-ERK/MMP axis is the most important molecular basis for the rapid switch towards tumorigenic conversion of the HrasA5 keratinocytes upon vemurafenib treatment. Finally, cotreatment with vemurafenib and the MEK inhibitor cobimetinib prevented MEK-ERK hyperactivation and with that abolished both, the epidermal differentiation and the tumor invasion phenotype. This suggests that both cutaneous adverse events are under direct control of vemurafenib-dependent MEK-ERK hyperactivation and confirms the dependence on preexisting genetic alterations of the skin keratinocytes that determine the basis towards induction of tumorigenic progression.

Download

Treatment resistance analysis reveals GLUT‐1‐mediated glucose uptake as a major target of synthetic rocaglates in cancer cells

September 2021

·

102 Reads

·

2 Citations

Rocaglates are natural compounds that have been extensively studied for their ability to inhibit translation initiation. Rocaglates represent promising drug candidates for tumor treatment due to their growth-inhibitory effects on neoplastic cells. In contrast to natural rocaglates, synthetic analogues of rocaglates have been less comprehensively characterized, but were also shown to have similar effects on the process of protein translation. Here, we demonstrate an enhanced growth-inhibitory effect of synthetic rocaglates when combined with glucose anti-metabolite 2-deoxy-D-glucose (2DG) in different cancer cell lines. Moreover, we unravel a new aspect in the mechanism of action of synthetic rocaglates involving reduction of glucose uptake mediated by downregulation or abrogation of glucose transporter GLUT-1 expression. Importantly, cells with genetically induced resistance to synthetic rocaglates showed substantially less pronounced treatment effect on glucose metabolism and did not demonstrate GLUT-1 downregulation, pointing at the crucial role of this mechanism for the anti-tumor activity of the synthetic rocaglates. Transcriptome profiling revealed glycolysis as one of the major pathways differentially regulated in sensitive and resistant cells. Analysis of synthetic rocaglate efficacy in a 3D tissue context with a co-culture of tumor and normal cells demonstrated a selective effect on tumor cells and substantiated the mechanistic observations obtained in cancer cell lines. Increased glucose uptake and metabolism is a universal feature across different tumor types. Therefore, targeting this feature by synthetic rocaglates could represent a promising direction for exploitation of rocaglates in novel anti-tumor therapies.


Organotypic Co-Cultures as a Novel 3D Model for Head and Neck Squamous Cell Carcinoma

August 2020

·

1,657 Reads

·

33 Citations

Background: Head and neck squamous cell carcinomas (HNSCC) are phenotypically and molecularly heterogeneous and frequently develop therapy resistance. Reliable patient-derived 3D tumor models are urgently needed to further study the complex pathogenesis of these tumors and to overcome treatment failure. Methods: We developed a three-dimensional organotypic co-culture (3D-OTC) model for HNSCC that maintains the architecture and cell composition of the individual tumor. A dermal equivalent (DE), composed of healthy human-derived fibroblasts and viscose fibers, served as a scaffold for the patient sample. DEs were co-cultivated with 13 vital HNSCC explants (non-human papillomavirus (HPV) driven, n = 7; HPV-driven, n = 6). Fractionated irradiation was applied to 5 samples (non-HPV-driven, n = 2; HPV-driven n = 3). To evaluate expression of ki-67, cleaved caspase-3, pan-cytokeratin, p16INK4a, CD45, ∝smooth muscle actin and vimentin over time, immunohistochemistry and immunofluorescence staining were performed Patient checkup data were collected for up to 32 months after first diagnosis. Results: All non-HPV-driven 3D-OTCs encompassed proliferative cancer cells during cultivation for up to 21 days. Proliferation indices of primaries and 3D-OTCs were comparable and consistent over time. Overall, tumor explants displayed heterogeneous growth patterns (i.e., invasive, expansive, silent). Cancer-associated fibroblasts and leukocytes could be detected for up to 21 days. HPV DNA was detectable in both primary and 3D-OTCs (day 14) of HPV-driven tumors. However, p16INK4a expression levels were varying. Morphological alterations and radioresistant tumor cells were detected in 3D-OTC after fractionated irradiation in HPV-driven and non-driven samples. Conclusions: Our 3D-OTC model for HNSCC supports cancer cell survival and proliferation in their original microenvironment. The model enables investigation of invasive cancer growth and might, in the future, serve as a platform to perform sensitivity testing upon treatment to predict therapy response.


Abstract 336: Organotypic co-cultures as a novel 3D model for head and neck squamous cell carcinoma

August 2020

·

16 Reads

Cancer Research

Background. Head and neck squamous cell carcinomas (HNSCC) are histologically and molecularly heterogeneous tumors and frequently develop resistance mechanisms against treatment. Reliable patient-derived 3D tumor models are urgently needed to further study the complex pathogenesis of these tumors and how to overcome treatment failure. Methods. We developed a 3D organotypic co-culture (OTC) model for HNSCCs that maintains the architecture and cell composition of the patients' tumor. A dermal equivalent, composed of healthy, human-derived fibroblasts embedded into viscose fibers, grows in an insert for approximately 10 days in culture and serves as a scaffold for the patient sample. After outgrowth of fibroblast within the dermal equivalent, we cultivated vital HNSCC tumor explants from 13 patients (non-HPV driven, n=7; HPV-driven HNSCC, n=6) by carefully placing them on top of the scaffold. Fractionated irradiation (IR) was applied to 5 samples. To evaluate tumor proliferation and areas of apoptosis, we performed immunohistochemistry and immunofluorescence with antibodies against ki-67, cleaved caspase-3, pan-cytokeratin (PanCK), p16INK4a, and vimentin. Results. All 3D OTC models of non-HPV driven samples encompassed proliferative cancer cells during cultivation for up to 21 days. Proliferation indices of primary and 3D-OTC models were comparable and consistent over time. Overall, tumor explants displayed heterogeneous growth and invasion patterns. Radioresistant and clonally expanding tumor cells were enriched in 3D-OTC models after fractionated IR. HPV DNA was detectable in both primary and OTC (day 14) of HPV-driven tumors; However, p16INK4a expression levels were varying. Conclusion. Our 3D-dimensional organotypic co-culture model for HNSCC supports cancer cell survival and proliferation in their original microenvironment. The model enables investigation of invasive cancer growth as well as clonal expansion of resistant cancer cells upon treatment. In the future, the introduced model might serve as a platform to perform sensitivity testing upon treatment to predict the patients' individual response to therapy. Citation Format: Julia C. Thierauf, Luca Engelmann, Natalia Koerich Laureano, Hans-Jürgen Stark, Elena-Sophie Prigge, Dominik Horn, Kolja Freier, Niels Grabe, Philippe Federspil, Peter K. Plinkert, Magnus von Knebel Doeberitz, Jochen Hess, Annette Affolter. Organotypic co-cultures as a novel 3D model for head and neck squamous cell carcinoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 336.


Fig. 3 Relation of HEVs and immune infiltration. Staining of the same tumour sample for CD20 (left), MECA-79 (middle) and CD3 (right) markers. CD20-positive cells clearly marked the germinal centre of the lymph follicle, whereas CD3-positive cells were co-localised with HEVs. Arrows indicate the examples of positive staining. Scale bars top to bottom CD20: 50 µm, 100 µm, 1 mm; MECA-79: 100 µm, 100 µm, 1 mm, 1 mm; CD3: 100 µm, 100 µm, 1 mm
Fig. 5 Graphical summary. Model of auto-immunisation loop in Lynch syndrome. In Lynch syndrome, the host immune system can be primed against FSP neoantigens by precursor lesions, MMR-deficient crypts, existing in the normal colonic mucosa long before tumour manifestation. Recurrent exposure of the immune cells to FSP neoantigens expressed in MMR-deficient crypts leads to the generation of an FSP-specific T-cell pool. When a tumour arises, FSP-specific T cells can promptly recognise the FSP neoantigens expressed on the tumour cells and release cytokines, which could induce neogenesis of HEVs. New HEVs, in turn, can recruit more naive T cells, which would undergo maturation through exposure to the neoantigens into FSP-specific T cells. Newly formed FSP-specific T cells release more cytokines, potentially leading to generation of more HEVs. This self-amplifying loop keeps MSI tumour cells under constant immune pressure and promotes selection and outgrowth of cell clones lacking antigen expression via MHC class I via acquisition of B2M mutations, enabling immune escape. Thereby, HEVs can contribute to immunoediting of MSI tumour cells.
High endothelial venules are associated with microsatellite instability, hereditary background and immune evasion in colorectal cancer

July 2019

·

112 Reads

·

25 Citations

British Journal of Cancer

Microsatellite-unstable (MSI) tumours show a high load of mutational neoantigens, as a consequence of DNA mismatch repair deficiency. Consequently, MSI tumours commonly present with dense immune infiltration and develop immune evasion mechanisms. Whether improved lymphocyte recruitment contributes to the pronounced immune infiltration in MSI tumours is unknown. We analysed the density of high endothelial venules (HEV) and postcapillary blood vessels specialised for lymphocyte trafficking, in MSI colorectal cancers (CRC). HEV density was determined by immunohistochemical staining of FFPE tissue sections from MSI (n = 48) and microsatellite-stable (MSS, n = 35) CRCs. Associations with clinical and pathological variables were analysed. We found elevated HEV densities in MSI compared with MSS CRCs (median 0.049 vs 0.000 counts/mm², respectively, p = 0.0002), with the highest densities in Lynch syndrome MSI CRCs. Dramatically elevated HEV densities were observed in B2M-mutant Lynch syndrome CRCs, pointing towards a link between lymphocyte recruitment and immune evasion (median 0.485 vs 0.0885 counts/mm² in B2M-wild-type tumours, p = 0.0237). Our findings for the first time indicate a significant contribution of lymphocyte trafficking in immune responses against MSI CRC, particularly in the context of Lynch syndrome. High HEV densities in B2M-mutant tumours underline the significance of immunoediting during tumour evolution.




Skin squamous cell carcinoma models: The role in combating the disease

January 2018

·

129 Reads

·

3 Citations

Nonmelanoma skin cancer is the most prevalent cancer worldwide, and a number of cases are steadily increasing. The incidence of skin squamous cell carcinoma, which is the second most frequent tumor in this group of tumors, is markedly increasing in immunosuppressed transplant recipients and under certain cancer treatment modalities. Unfortunately, the aggressiveness of these tumors is also increasing, and once metastasized, the therapies to successfully eradicate these tumors are lacking, thus making research advancements vital. We firstly discuss what is currently known about cutaneous squamous cell carcinomas (cSCCs), an important consideration when designing relevant model systems for further investigation. We continue by reviewing recent studies, which have utilized different models and highlight the importance/requirement for human 3D models to combat this devastating tumor type.



Citations (49)


... Despite numerous attempts, restoring the thymic function has been challenging due to the difficulty to reproduce the thymic microenvironment. Notably, thymic three-dimensional (3-D) configuration is critical to the survival and function of TECs, whereas TECs cultured in a 2-D environment rapidly decrease the expression of genes critical for thymopoiesis 3,4 . ...

Reference:

Promoting 3-D aggregation of FACS purified thymic epithelial cells with EAK 16-II/EAKIIH6 self-assembling hydrogel
3D Organotypic Co-culture Model Supporting Medullary Thymic Epithelial Cell Proliferation, Differentiation and Promiscuous Gene Expression
  • Citing Article
  • July 2015

Journal of Visualized Experiments

... 23 The activation of the RAS−RAF−MEK−ERK signaling pathway is an important molecular basis for the skin disease and cure. 24 When skin is wounded, the Ras/Raf/ERK/ MEK signaling pathway is also obviously damaged. 25 Skin wound is a strong stimulus, which can be used as a signal to activate the Ras/Raf/MEK/ERK signaling pathway. ...

Adverse Effects of Vemurafenib on Skin Integrity: Hyperkeratosis and Skin Cancer Initiation Due to Altered MEK/ERK-Signaling and MMP Activity

... It was first evaluated as an anti-Parkinson disease agent, demonstrating a promising activity and potential for developing novel drugs [79]. However, it was recently tested with the analog IMD-026260 (structure VIII in Fig. 5) against colorectal cancer cells [80]. The synthetic procedure for IMD-026260 has been described by Diedrichs et al. [81] and is depicted in Fig. 4. IMD-026259 and IMD-026260 cause cancer cell death by inhibiting glucose metabolism and absorption thereof, which is mediated by the regulation of the glucose transporter-1 (GLUT-1), presenting for the first time the importance of regulation of GLUT-1 as a prime target for synthetic rocaglates [80]. ...

Treatment resistance analysis reveals GLUT‐1‐mediated glucose uptake as a major target of synthetic rocaglates in cancer cells

... The applicability and feasibility of this 3D organotypic model are expected to increase for various oral cancer studies. Nevertheless, this model presents basic disadvantages due to the manual workflow, like any organotypic culture model, including the vital explant co-culture model [53]. It includes low throughput, a lack of reproducibility, limited scalability, and skillful handling. ...

Organotypic Co-Cultures as a Novel 3D Model for Head and Neck Squamous Cell Carcinoma

... In contrast to their absence in healthy colonic tissue, HEVs have been identified at the periphery of tumor infiltrates in CRC, where their presence is indicative of an adverse prognosis [76,77]. Although they are distributed differently, HEV in the tumor and peritumor areas have no morphological differences [78]. HEV is also a prognostic biomarker in CRC. ...

High endothelial venules are associated with microsatellite instability, hereditary background and immune evasion in colorectal cancer

British Journal of Cancer

... Immunosuppression is also a very relevant risk factor for squamous cell carcinoma (SCC), as immunosuppressed transplant recipients bear a 100-fold risk for the development of cutaneous SCC. In these cases, the tumors occur earlier in life and are more aggressive, with a metastasis rate of up to 10% [37]. Cutaneous human papillomavirus of the genus beta is a particular risk factor in SCC. ...

Skin squamous cell carcinoma models: The role in combating the disease
  • Citing Chapter
  • January 2018

... In addition, GDF11 is also associated with the inactivation of inflammatory responses and protection against them by inhibiting oxidative stress and the expression of heat shock proteins [47]. Downregulation of homeodomain interaction protein kinase 4 was determined in our analysis; its silencing was found to be related to skin epithelial differentiation from stem cells in a previous study [48]. ...

An RNAi Screen Reveals an Essential Role for HIPK4 in Human Skin Epithelial Differentiation from iPSCs

Stem Cell Reports

... The highest collagen content was found in males older than 40, whereas for women in a 65-year-old donor and some subjects younger than 30 years old, which contradicts the notion of decreased collagen expression in in-vivo aged skin. Kaisers et al. also showed that the changes in gene expression of in-vivo aged skin are not represented in short-term cultures [55]. Furthermore, in the unpublished study mentioned above, we have also seen a 75% increase in COL1A1 gene expression in the older age group compared to the younger group. ...

Age, gender and UV-exposition related effects on gene expression in in vivo aged short term cultivated human dermal fibroblasts

... Studies on keratinocytes have been greatly facilitated by the fact that these cells can be isolated from mammalian skin and can be easily grown in culture, where they retain much of their in vivo properties [1]. Keratinocytes in culture exhibit polarity, grow as epithelial sheets and can be induced to undergo terminal squamous differentiation [2,3,4]. Therefore, these cells have been widely utilized to study various biological processes of epithelial cells such as cell cycle progression, differentiation, migration, and wound healing [4,5]. ...

Experimental Models to Analyze Differentiation Functions of Cultured Keratinocytes In Vitro and In Vivo
  • Citing Chapter
  • January 2004

... A more sophisticated approach using 3D scaffolds offers a promising strategy to imitate the thymic extracellular matrix in 3D thymic co-culture models. Artificially generated matrices consisting of fibrin gel mixed with fibroblasts have been reported to promote the proliferation, differentiation, and functionality of TECs [76]. In a later study, a 3D scaffold was directly generated from decellularized thymic tissues and proven suitable for the long-term in vitro cultivation of TECs with functional thymopoiesis [77]. ...

3D Organotypic Co-culture Model Supporting Medullary Thymic Epithelial Cell Proliferation, Differentiation and Promiscuous Gene Expression
  • Citing Article
  • July 2015

Journal of Visualized Experiments