Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Science ; 384(6699): eadi7453, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38815020

ABSTRACT

Stem cells play a critical role in cancer development by contributing to cell heterogeneity, lineage plasticity, and drug resistance. We created gene expression networks from hundreds of mouse tissue samples (both normal and tumor) and integrated these with lineage tracing and single-cell RNA-seq, to identify convergence of cell states in premalignant tumor cells expressing markers of lineage plasticity and drug resistance. Two of these cell states representing multilineage plasticity or proliferation were inversely correlated, suggesting a mutually exclusive relationship. Treatment of carcinomas in vivo with chemotherapy repressed the proliferative state and activated multilineage plasticity whereas inhibition of differentiation repressed plasticity and potentiated responses to cell cycle inhibitors. Manipulation of this cell state transition point may provide a source of potential combinatorial targets for cancer therapy.


Subject(s)
Carcinoma, Squamous Cell , Cell Lineage , Neoplastic Stem Cells , Skin Neoplasms , Animals , Mice , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Neoplastic Stem Cells/pathology , Single-Cell Analysis , Cell Differentiation , Drug Resistance, Neoplasm/genetics , Cell Plasticity , Cell Proliferation , Gene Regulatory Networks , RNA-Seq , Gene Expression Regulation, Neoplastic
2.
bioRxiv ; 2023 May 10.
Article in English | MEDLINE | ID: mdl-37215032

ABSTRACT

Adult mammalian stem cells play critical roles in normal tissue homeostasis, as well as in tumor development, by contributing to cell heterogeneity, plasticity, and development of drug resistance. The relationship between different types of normal and cancer stem cells is highly controversial and poorly understood. Here, we carried out gene expression network analysis of normal and tumor samples from genetically heterogeneous mice to create network metagenes for visualization of stem-cell networks, rather than individual stem-cell markers, at the single-cell level during multistage carcinogenesis. We combined this approach with lineage tracing and single-cell RNASeq of stem cells and their progeny, identifying a previously unrecognized hierarchy in which Lgr6+ stem cells from tumors generate progeny that express a range of other stem-cell markers including Sox2, Pitx1, Foxa1, Klf5, and Cd44. Our data identify a convergence of multiple stem-cell and tumor-suppressor pathways in benign tumor cells expressing markers of lineage plasticity and oxidative stress. This same single-cell population expresses network metagenes corresponding to markers of cancer drug resistance in human tumors of the skin, lung and prostate. Treatment of mouse squamous carcinomas in vivo with the chemotherapeutic cis-platin resulted in elevated expression of the genes that mark this cell population. Our data have allowed us to create a simplified model of multistage carcinogenesis that identifies distinct stem-cell states at different stages of tumor progression, thereby identifying networks involved in lineage plasticity, drug resistance, and immune surveillance, providing a rich source of potential targets for cancer therapy.

3.
Nat Cell Biol ; 20(6): 699-709, 2018 06.
Article in English | MEDLINE | ID: mdl-29802408

ABSTRACT

Tumour cells are subjected to evolutionary selection pressures during progression from initiation to metastasis. We analysed the clonal evolution of squamous skin carcinomas induced by DMBA/TPA treatment using the K5CreER-Confetti mouse and stage-specific lineage tracing. We show that benign tumours are polyclonal, but only one population contains the Hras driver mutation. Thus, benign papillomas are monoclonal in origin but recruit neighbouring epithelial cells during growth. Papillomas that never progress to malignancy retain several distinct clones, whereas progression to carcinoma is associated with a clonal sweep. Newly generated clones within carcinomas demonstrate intratumoural invasion and clonal intermixing, often giving rise to metastases containing two or more distinct clones derived from the matched primary tumour. These data demonstrate that late-stage tumour progression and dissemination are governed by evolutionary selection pressures that operate at a multicellular level and, therefore, differ from the clonal events that drive initiation and the benign-malignant transition.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Lineage , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Clonal Evolution , Epithelial Cells/pathology , Neoplasms, Experimental/genetics , Skin Neoplasms/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Cell Proliferation/genetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, ras , Genetic Predisposition to Disease , Male , Mice, Transgenic , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phenotype , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate , Time Factors , Tumor Burden/genetics
4.
Elife ; 62017 12 04.
Article in English | MEDLINE | ID: mdl-29199946

ABSTRACT

The cutaneous wound-healing program is a product of a complex interplay among diverse cell types within the skin. One fundamental process that is mediated by these reciprocal interactions is the mobilization of local stem cell pools to promote tissue regeneration and repair. Using the ablation of epidermal caspase-8 as a model of wound healing in Mus musculus, we analyzed the signaling components responsible for epithelial stem cell proliferation. We found that IL-1α and IL-7 secreted from keratinocytes work in tandem to expand the activated population of resident epidermal γδT-cells. A downstream effect of activated γδT-cells is the preferential proliferation of hair follicle stem cells. By contrast, IL-1α-dependent stimulation of dermal fibroblasts optimally stimulates epidermal stem cell proliferation. These findings provide new mechanistic insights into the regulation and function of epidermal cell-immune cell interactions and into how components that are classically associated with inflammation can differentially influence distinct stem cell niches within a tissue.


Subject(s)
Cell Proliferation , Hair Follicle/cytology , Interleukin-1alpha/metabolism , Intraepithelial Lymphocytes/physiology , Stem Cells/physiology , Wound Healing , Animals , Interleukin-7/metabolism , Lymphocyte Activation , Mice , Models, Animal
5.
Nat Genet ; 49(11): 1624-1632, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945253

ABSTRACT

The G-protein-coupled receptors LGR4, LGR5 and LGR6 are Wnt signaling mediators, but their functions in squamous cell carcinoma (SCC) are unclear. Using lineage tracing in Lgr5-EGFP-CreERT2/Rosa26-Tomato and Lgr6-EGFP-CreERT2/Rosa26-Tomato reporter mice, we demonstrate that Lgr6, but not Lgr5, acts as an epithelial stem cell marker in SCCs in vivo. We identify, by single-molecule in situ hybridization and cell sorting, rare cells positive for Lgr6 expression in immortalized keratinocytes and show that their frequency increases in advanced SCCs. Lgr6 expression is enriched in cells with stem cell characteristics, and Lgr6 downregulation in vivo causes increased epidermal proliferation with expanded lineage tracing from epidermal stem cells positive for Lgr6 expression. Surprisingly, mice with germline knockout of Lgr6 are predisposed to SCC development, through a mechanism that includes compensatory upregulation of Lgr5. These data provide a model for human patients with germline loss-of-function mutations in Wnt pathway genes, including RSPO1 or LGR4, who show increased susceptibility to squamous tumor development.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Keratinocytes/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Skin Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Transformed , Epidermis/metabolism , Epidermis/pathology , Humans , Keratinocytes/pathology , Mice , Mice, Transgenic , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thrombospondins/genetics , Thrombospondins/metabolism
6.
Elife ; 62017 07 11.
Article in English | MEDLINE | ID: mdl-28695824

ABSTRACT

The control principles behind robust cyclic regeneration of hair follicles (HFs) remain unclear. Using multi-scale modeling, we show that coupling inhibitors and activators with physical growth of HFs is sufficient to drive periodicity and excitability of hair regeneration. Model simulations and experimental data reveal that mouse skin behaves as a heterogeneous regenerative field, composed of anatomical domains where HFs have distinct cycling dynamics. Interactions between fast-cycling chin and ventral HFs and slow-cycling dorsal HFs produce bilaterally symmetric patterns. Ear skin behaves as a hyper-refractory domain with HFs in extended rest phase. Such hyper-refractivity relates to high levels of BMP ligands and WNT antagonists, in part expressed by ear-specific cartilage and muscle. Hair growth stops at the boundaries with hyper-refractory ears and anatomically discontinuous eyelids, generating wave-breaking effects. We posit that similar mechanisms for coupled regeneration with dominant activator, hyper-refractory, and wave-breaker regions can operate in other actively renewing organs.


Subject(s)
Hair Follicle/physiology , Hair/growth & development , Animals , Mice , Models, Biological , Regeneration , Spatio-Temporal Analysis
7.
Cell Rep ; 16(4): 1153-1165, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27425619

ABSTRACT

Inherited germline polymorphisms can cause gene expression levels in normal tissues to differ substantially between individuals. We present an analysis of the genetic architecture of normal adult skin from 470 genetically unique mice, demonstrating the effect of germline variants, skin tissue location, and perturbation by exogenous inflammation or tumorigenesis on gene signaling pathways. Gene networks related to specific cell types and signaling pathways, including sonic hedgehog (Shh), Wnt, Lgr family stem cell markers, and keratins, differed at these tissue sites, suggesting mechanisms for the differential susceptibility of dorsal and tail skin to development of skin diseases and tumorigenesis. The Pten tumor suppressor gene network is rewired in premalignant tumors compared to normal tissue, but this response to perturbation is lost during malignant progression. We present a software package for expression quantitative trait loci (eQTL) network analysis and demonstrate how network analysis of whole tissues provides insights into interactions between cell compartments and signaling molecules.


Subject(s)
Carcinogenesis/genetics , Gene Expression/genetics , Inflammation/genetics , Inflammation/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin/pathology , Animals , Carcinogenesis/pathology , Disease Progression , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Germ Cells/physiology , Mice , Polymorphism, Genetic/genetics , Quantitative Trait Loci/genetics , Signal Transduction/genetics
8.
Proc Natl Acad Sci U S A ; 111(42): 15114-9, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25277970

ABSTRACT

Regulation of adult stem cells (SCs) is fundamental for organ maintenance and tissue regeneration. On the body surface, different ectodermal organs exhibit distinctive modes of regeneration and the dynamics of their SC homeostasis remain to be unraveled. A slow cycling characteristic has been used to identify SCs in hair follicles and sweat glands; however, whether a quiescent population exists in continuously growing nails remains unknown. Using an in vivo label retaining cells (LRCs) system, we detected an unreported population of quiescent cells within the basal layer of the nail proximal fold, organized in a ring-like configuration around the nail root. These nail LRCs express the hair stem cell marker, keratin 15 (K15), and lineage tracing show that these K15-derived cells can contribute to both the nail structure and peri-nail epidermis, and more toward the latter. Thus, this stem cell population is bifunctional. Upon nail plucking injury, the homeostasis is tilted with these SCs dominantly delivering progeny to the nail matrix and differentiated nail plate, demonstrating their plasticity to adapt to wounding stimuli. Moreover, in vivo engraftment experiments established that transplanted nail LRCs can actively participate in functional nail regeneration. Transcriptional profiling of isolated nail LRCs revealed bone morphogenetic protein signaling favors nail differentiation over epidermal fate. Taken together, we have found a previously unidentified ring-configured population of bifunctional SCs, located at the interface between the nail appendage organ and adjacent epidermis, which physiologically display coordinated homeostatic dynamics but are capable of rediverting stem cell flow in response to injury.


Subject(s)
Ectoderm/cytology , Hoof and Claw/cytology , Regeneration , Stem Cells/cytology , Wound Healing , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Epidermal Cells , Green Fluorescent Proteins/metabolism , Homeostasis , Mice , Mice, Knockout , Signal Transduction , Transcription, Genetic
9.
Stem Cells ; 32(8): 2267-77, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24715701

ABSTRACT

Adult stem cells (SCs) are important to maintain homeostasis of tissues including several mini-organs like hair follicles and sweat glands. However, the existence of stem cells in minor salivary glands (SGs) is largely unexplored. In vivo histone2B green fluorescent protein pulse chase strategy has allowed us to identify slow-cycling, label-retaining cells (LRCs) of minor SGs that preferentially localize in the basal layer of the lower excretory duct with a few in the acini. Engraftment of isolated SG LRC in vivo demonstrated their potential to differentiate into keratin 5 (basal layer marker) and keratin 8 (luminal layer marker)-positive structures. Transcriptional analysis revealed activation of TGFß1 target genes in SG LRC and BMP signaling in SG progenitors. We also provide evidence that minor SGSCs are sensitive to tobacco-derived tumor-inducing agent and give rise to tumors resembling low grade adenoma. Our data highlight for the first time the existence of minor SG LRCs with stem cells characteristic and emphasize the role of transforming growth factor beta (TGFß) pathway in their maintenance.


Subject(s)
Adult Stem Cells/cytology , Cell Separation/methods , Salivary Glands, Minor/cytology , Animals , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Stem Cells ; 32(2): 534-47, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24023003

ABSTRACT

Hair follicles (HFs) are regenerative miniorgans that offer a highly informative model system to study the regulatory mechanisms of hair follicle stem cells (hfSCs) homeostasis and differentiation. Bone morphogenetic protein (BMP) signaling is key in both of these processes, governing hfSCs quiescence in the bulge and differentiation of matrix progenitors. However, whether canonical or noncanonical pathways of BMP signaling are responsible for these processes remains unresolved. Here, we conditionally ablated two canonical effectors of BMP signaling, Smad1 and Smad5 during hair morphogenesis and postnatal cycling in mouse skin. Deletion of Smad1 and Smad5 (dKO) in the epidermis during morphogenesis resulted in neonatal lethality with lack of visible whiskers. Interestingly, distinct patterns of phospho-Smads (pSmads) activation were detected with pSmad8 restricted to epidermis and pSmad1 and pSmad5 exclusively activated in HFs. Engraftment of dKO skin revealed retarded hair morphogenesis and failure to differentiate into visible hair. The formation of the prebulge and bulge reservoir for quiescent hfSCs was precluded in dKO HFs which remained in prolonged anagen. Surprisingly, in postnatal telogen HFs, pSmad8 expression was no longer limited to epidermis and was also present in dKO bulge hfSCs and matrix progenitors. Although pSmad8 activity alone could not prevent dKO hfSCs precocious anagen activation, it sustained efficient postnatal differentiation and regeneration of visible hairs. Together, our data suggest a pivotal role for canonical BMP signaling demonstrating distinguished nonoverlapping function of pSmad8 with pSmad1 and pSmad5 in hfSCs regulation and hair morphogenesis but a redundant role in adult hair progenitors differentiation.


Subject(s)
Hair Follicle/growth & development , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Animals , Cell Differentiation , Epidermis/growth & development , Epidermis/metabolism , Hair/growth & development , Hair/metabolism , Hair Follicle/metabolism , Mice , Morphogenesis/genetics , Regeneration , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics , Stem Cells/metabolism
11.
Stem Cells ; 32(4): 886-901, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24222445

ABSTRACT

The hair follicle (HF) is an exceptional mini-organ to study the mechanisms which regulate HF morphogenesis, cycling, hair follicle stem cell (hfSCs) homeostasis, and progeny differentiation. During morphogenesis, Wnt signaling is well-characterized in the initiation of HF patterning but less is known about which particular Wnt ligands are required and whether individual Wnt ligands act in an indispensable or redundant manner during postnatal hfSCs anagen onset and HF cycle progression. Previously, we described the function of the bone morphogenetic protein (BMP) signaling target gene WNT7a in intrinsic regulation of hfSCs homeostasis in vivo. Here, we investigated the role of Wnt7b, which was also intrinsically upregulated in hfSCs during physiological and precocious anagen after BMP inhibition in vivo. We demonstrated Wnt7b to be a direct target of canonical BMP signaling in hfSCs and using Wnt7b conditional gene targeting during HF morphogenesis revealed disrupted HF cycling including a shorter anagen, premature catagen onset with overall shorter hair production, and diminished HF differentiation marker expression. Additionally, we observed that postnatal ablation of Wnt7b resulted in delayed HF activation, affecting both the hair germ and bulge hfSCs but still maintaining a two-step sequence of HF stimulation. Interestingly, Wnt7b cKO hfSCs participated in reformation of the new HF bulge, but with slower self-renewal. These findings demonstrate the importance of intrinsic Wnt7b expression in hfSCs regulation and normal HF cycling and surprisingly reveal a nonredundant role for Wnt7b in the control of HF anagen length and catagen entry which was not compensated by other Wnt ligands.


Subject(s)
Hair Follicle/metabolism , Homeostasis/physiology , Proto-Oncogene Proteins/metabolism , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation/physiology , Hair Follicle/cytology , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Stem Cells/cytology , Wnt Proteins/genetics
12.
PLoS One ; 8(9): e74174, 2013.
Article in English | MEDLINE | ID: mdl-24058524

ABSTRACT

Slow cycling is a common feature shared among several stem cells (SCs) identified in adult tissues including hair follicle and cornea. Recently, existence of unipotent SCs in basal and lumenal layers of sweat gland (SG) has been described and label retaining cells (LRCs) have also been localized in SGs; however, whether these LRCs possess SCs characteristic has not been investigated further. Here, we used a H2BGFP LRCs system for in vivo detection of infrequently dividing cells. This system allowed us to specifically localize and isolate SCs with label-retention and myoepithelial characteristics restricted to the SG proximal acinar region. Using an alternative genetic approach, we demonstrated that SG LRCs expressed keratin 15 (K15) in the acinar region and lineage tracing determined that K15 labeled cells contributed long term to the SG structure but not to epidermal homeostasis. Surprisingly, wound healing experiments did not activate proximal acinar SG cells to participate in epidermal healing. Instead, predominantly non-LRCs in the SG duct actively divided, whereas the majority of SG LRCs remained quiescent. However, when we further challenged the system under more favorable isolated wound healing conditions, we were able to trigger normally quiescent acinar LRCs to trans-differentiate into the epidermis and adopt its long term fate. In addition, dissociated SG cells were able to regenerate SGs and, surprisingly, hair follicles demonstrating their in vivo plasticity. By determining the gene expression profile of isolated SG LRCs and non-LRCs in vivo, we identified several Bone Morphogenetic Protein (BMP) pathway genes to be up-regulated and confirmed a functional requirement for BMP receptor 1A (BMPR1A)-mediated signaling in SG formation. Our data highlight the existence of SG stem cells (SGSCs) and their primary importance in SG homeostasis. It also emphasizes SGSCs as an alternative source of cells in wound healing and their plasticity for regenerating different skin appendages.


Subject(s)
Epidermal Cells , Epithelial Cells/cytology , Hair Follicle/cytology , Stem Cells/cytology , Sweat Glands/cytology , Wound Healing/genetics , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Epidermis/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Follicle/metabolism , Keratin-15/genetics , Keratin-15/metabolism , Mice , Mice, Nude , Mice, Transgenic , Regeneration , Signal Transduction , Stem Cells/metabolism , Sweat Glands/metabolism
13.
Proc Natl Acad Sci U S A ; 110(4): 1351-6, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23292934

ABSTRACT

Hair follicles facilitate the study of stem cell behavior because stem cells in progressive activation stages, ordered within the follicle architecture, are capable of cyclic regeneration. To study the gene network governing the homeostasis of hair bulge stem cells, we developed a Keratin 15-driven genetic model to directly perturb molecular signaling in the stem cells. We visualize the behavior of these modified stem cells, evaluating their hair-regenerating ability and profile their molecular expression. Bone morphogenetic protein (BMP)-inactivated stem cells exhibit molecular profiles resembling those of hair germs, yet still possess multipotentiality in vivo. These cells also exhibit up-regulation of Wnt7a, Wnt7b, and Wnt16 ligands and Frizzled (Fzd) 10 receptor. We demonstrate direct transcriptional modulation of the Wnt7a promoter. These results highlight a previously unknown intra-stem cell antagonistic competition, between BMP and Wnt signaling, to balance stem cell activity. Reduced BMP signaling and increased Wnt signaling tilts each stem cell toward a hair germ fate and, vice versa, based on a continuous scale dependent on the ratio of BMP/Wnt activity. This work reveals one more hierarchical layer regulating stem cell homeostasis beneath the stem cell-dermal papilla-based epithelial-mesenchymal interaction layer and the hair follicle-intradermal adipocyte-based tissue interaction layer. Although hierarchical layers are all based on BMP/Wnt signaling, the multilayered control ensures that all information is taken into consideration and allows hair stem cells to sum up the total activators/inhibitors involved in making the decision of activation.


Subject(s)
Adult Stem Cells/metabolism , Bone Morphogenetic Proteins/metabolism , Gene Regulatory Networks , Hair Follicle/cytology , Hair Follicle/metabolism , Wnt Signaling Pathway , Adult Stem Cells/cytology , Animals , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Hair/growth & development , Hair Follicle/drug effects , Homeostasis/genetics , Keratin-15/genetics , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Models, Genetic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Promoter Regions, Genetic , Transcriptome , Wnt Proteins/administration & dosage
14.
Methods Mol Biol ; 585: 303-12, 2010.
Article in English | MEDLINE | ID: mdl-19908012

ABSTRACT

Three-dimensional (3D) organotypic models are increasingly being used to study aspects of epidermal organisation and cutaneous wound-healing events. These are largely dependent on laborious histological analysis and immunohistochemical approaches. Here we outline a method for establishment of a versatile in vitro 3D organotypic skin equivalent that reflects murine epidermal organisation in vivo. The system is optically transparent and ideally suited to real-time analysis and integrated in situ imaging techniques. Moreover, the model permits the visualisation of epidermal regeneration following injury in real time, thereby facilitating avenues to explore distinctive modes of wound re-epithelialisation. The versatility of the model could help unravel molecular mechanisms underlying epidermal morphogenesis, assess novel therapeutic strategies and reduce animal experimentation in a non-invasive manner.


Subject(s)
Epidermis/physiology , Wound Healing/physiology , Animals , Mice , Regeneration , Tissue Engineering
15.
J Invest Dermatol ; 128(4): 1039-49, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17960178

ABSTRACT

Three-dimensional (3D) organotypic models are increasingly used to study the aspects of epidermal organisation and cutaneous wound-healing events. However, these are largely dependent on laborious histological analysis and immunohistochemical approaches. Despite the large resource of transgenic and knockout mice harboring mutations relevant to skin disorders, few organotypic mouse skin models are available. We have developed a versatile in vitro 3D organotypic mouse skin equivalent that reflects epidermal organisation in vivo. The system is optically transparent and ideally suited to real-time analysis using a variety of integrated in situ imaging techniques. As a paradigm for coordination of cellular events, the epidermal gap junction network was investigated and the model displayed predominant connexin 43 (Cx43) expression in basal proliferating cells and Cx26 and Cx30 expression in differentiated keratinocytes. We show that attenuation of Cx43-mediated communication by a Cx mimetic peptide enhanced wound closure rates in keratinocyte monocultures and in the living skin equivalent system, emphasising the utility of the model to systematically unravel the molecular mechanisms underlying epidermal morphogenesis, assess promising therapeutic strategies, and reduce animal experimentation. Furthermore, we visualise epidermal regeneration following injury in real time, thereby facilitating avenues to explore distinctive modes of wound re-epithelialisation in a non-invasive manner.


Subject(s)
Connexins/physiology , Epidermal Cells , Epidermis/physiology , Organ Culture Techniques/methods , Animals , Connexin 26 , Connexin 30 , Connexin 43/analysis , Connexin 43/physiology , Connexins/analysis , Epidermis/drug effects , Gap Junctions , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/physiology , Mice , Regeneration , Skin/cytology , Skin/drug effects , Wound Healing/drug effects
16.
Liver Int ; 25(2): 389-402, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780065

ABSTRACT

BACKGROUND: Liver cell lines closely resembling primary hepatocyte are essential for research on hepatitis viruses and hepatocyte function. Currently used cell lines are derived from hepatic tumours and have altered gene expression. AIMS: The generation and characterisation of novel human hepatocyte lines (HHLs) derived from healthy human liver, retaining the primary hepatocyte phenotype. RESULTS: Primary hepatocytes were immortalised with Moloney's mouse leukaemia virus expressing E6 and E7 proteins of human papillomavirus, and cultures propagated long-term. All HHLs contained markers of hepatocyte and biliary phenotype (cytokeratins 7, 8, 18 and 19), Cytochrome P450 and albumin. The HHLs did not express high levels of p53 or alpha-fetoprotein. When grown in a collagen sandwich culture, or at the air-liquid interface, HHLs were maintained as monolayer whereas Huh-7 and HepG2 formed thick layers. All HHLs showed increased capacity to bind recombinant hepatitis C virus-like particles in comparison with Huh-7 and HepG2. We also demonstrate that HHLs contained active gap junctions, and that the cells respond to stimulation with IFN-alpha by upregulation of major histocompatibility complex (MHC)-I and -II. CONCLUSIONS: These HHLs retain primary hepatocyte phenotype and should be useful for investigating mechanisms of entry and replication of hepatotropic viruses, and should also be valuable in the study of hepatocyte biology and pathology.


Subject(s)
Cell Communication/immunology , Hepatocytes/cytology , Hepatocytes/immunology , Interferon-gamma/pharmacology , Binding Sites , Cell Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Gap Junctions , Hepatocytes/drug effects , Humans , Male , Reference Values , Sampling Studies , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...