Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Hum Genet ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38815586

ABSTRACT

To date, clinical genetic testing for Mendelian disease variants has focused heavily on exonic coding and intronic gene regions. This multi-step study was undertaken to provide an evidence base for selecting and applying computational approaches for use in clinical classification of 5' cis-regulatory region variants. Curated datasets of clinically reported disease-causing 5' cis-regulatory region variants and variants from matched genomic regions in population controls were used to calibrate six bioinformatic tools as predictors of variant pathogenicity. Likelihood ratio estimates were aligned to code weights following ClinGen recommendations for application of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) classification scheme. Considering code assignment across all reference dataset variants, performance was best for CADD (81.2%) and REMM (81.5%). Optimized thresholds provided moderate evidence toward pathogenicity (CADD, REMM) and moderate (CADD) or supporting (REMM) evidence against pathogenicity. Both sensitivity and specificity of prediction were improved when further categorizing variants based on location in an EPDnew-defined promoter region. Combining predictions (CADD, REMM, and location in a promoter region) increased specificity at the expense of sensitivity. Importantly, the optimal CADD thresholds for assigning ACMG/AMP codes PP3 (≥10) and BP4 (≤8) were vastly different from recommendations for protein-coding variants (PP3 ≥25.3; BP4 ≤22.7); CADD <22.7 would incorrectly assign BP4 for >90% of reported disease-causing cis-regulatory region variants. Our results demonstrate the need to consider a tiered approach and tailored score thresholds to optimize bioinformatic impact prediction for clinical classification of 5' cis-regulatory region variants.

2.
Exp Dermatol ; 29(5): 450-461, 2020 05.
Article in English | MEDLINE | ID: mdl-32145039

ABSTRACT

Mouse dorsal coat hair types, guard, awl, auchene and zigzag, develop in three consecutive waves. To date, it is unclear if these hair types are determined genetically through expression of specific factors or can change based on their mesenchymal environment. We undertook a novel approach to this question by studying individual hair type in 67 Collaborative Cross (CC) mouse lines and found significant variation in the proportion of each type between strains. Variation in the proportion of zigzag, awl and auchene, but not guard hair, was largely due to germline genetic variation. We utilised this variation to map a quantitative trait locus (QTL) on chromosome 12 that appears to influence a decision point switch controlling the propensity for either second (awl and auchene) or third wave (zigzag) hairs to develop. This locus contains two strong candidates, Sostdc1 and Twist1, each of which carry several ENCODE regulatory variants, specific to the causal allele, that can influence gene expression, are expressed in the developing hair follicle, and have been previously reported to be involved in regulating human and murine hair behaviour, but not hair subtype determination. Both of these genes are likely to play a part in hair type determination via regulation of BMP and/or WNT signalling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Chromosome Mapping , Genome-Wide Association Study , Hair/physiology , Polymorphism, Genetic , Signal Transduction , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Crosses, Genetic , Dermis/metabolism , Genetic Linkage , Mice , Phenotype , Quantitative Trait Loci , Species Specificity , Twist-Related Protein 1/genetics
3.
J Exp Clin Cancer Res ; 37(1): 266, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30382874

ABSTRACT

Most chemotherapeutics elevate intracellular levels of reactive oxygen species (ROS), and many can alter redox-homeostasis of cancer cells. It is widely accepted that the anticancer effect of these chemotherapeutics is due to the induction of oxidative stress and ROS-mediated cell injury in cancer. However, various new therapeutic approaches targeting intracellular ROS levels have yielded mixed results. Since it is impossible to quantitatively detect dynamic ROS levels in tumors during and after chemotherapy in clinical settings, it is of increasing interest to apply mathematical modeling techniques to predict ROS levels for understanding complex tumor biology during chemotherapy. This review outlines the current understanding of the role of ROS in cancer cells during carcinogenesis and during chemotherapy, provides a critical analysis of the methods used for quantitative ROS detection and discusses the application of mathematical modeling in predicting treatment responses. Finally, we provide insights on and perspectives for future development of effective therapeutic ROS-inducing anticancer agents or antioxidants for cancer treatment.


Subject(s)
Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Humans , Neoplasms/pathology , Oxidative Stress
4.
J Invest Dermatol ; 134(6): 1519-1526, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24531689

ABSTRACT

Hair follicles (HFs) upon development enter a lifelong cycle of growth, regression, and resting. These phases have been extensively studied at the cellular and molecular levels for individual HFs. However, HFs group into domains with coordinated cycling strongly influenced by their environment. These macroscopic hair domains have been difficult to study and can be influenced by physiological or pathological conditions, such as pregnancy or skin wounds. To robustly address this issue, we generated a mouse model for quantitative monitoring of ß-catenin activity reflecting HF cycle dynamics macroscopically by using live bioluminescence imaging. These mice allowed live tracking of HF cycles and development, and highlighted hair regenerative patterns known to occur through macro-environmental cues, including initiation events, propagating anagen and border stability, and allowed refinement of a mechanistic mathematical model that integrates epidermal cell population dynamics into an excitable reaction-diffusion model. HF cycling could be studied in situations of pregnancy, wound healing, hair plucking, as well as in response to cyclosporine or Wnt3a stimulation. In conclusion, we developed a model for analysis of HF cycling at the macroscopic level that will allow refined analysis of hair cycle kinetics as well as its propagation dynamics.


Subject(s)
Hair Follicle/growth & development , Hair/embryology , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cyclosporine/chemistry , Female , Genes, Reporter , Hair/physiology , Hair Follicle/metabolism , Luciferases, Firefly/genetics , Luminescence , Mice , Mice, Transgenic , Models, Theoretical , Pregnancy , Transgenes , Wnt3A Protein/metabolism , Wound Healing
5.
J Invest Dermatol ; 134(7): 1991-1997, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24509534

ABSTRACT

Epidemiological studies suggest that ultraviolet B exposure (UVR) during childhood is the most important environmental risk factor for melanoma. In accordance, neonatal, but not adult, UVR exacerbates melanoma incidence in mouse models. The inability of neonates, as opposed to adults, to mount a proper neutrophil inflammatory response in the skin upon UVR exposure has been one of the driving hypotheses explaining this observation for the past decade. However, this aspect remains controversial. Here, we evaluated the UVR-induced inflammatory response in neonatal versus adult mice. In neonates, a significant neutrophil infiltration could be identified and quantified using three different antibodies by flow cytometry or immunohistochemistry. On day 1 after UVR, neutrophils were increased by 84-fold and on day 4 macrophages increased by 37-fold compared with nonexposed age-matched skin. When compared with adults, neonatal skin harbored a higher proportion of neutrophils in the myeloid compartment without significant differences in absolute counts. This response was reproduced with different kinetics in C57Bl/6 and FVB mice with a more rapid attenuation of neutrophil counts in the latter. Overall, our results suggest that the greatly increased sensitivity to melanomagenesis in neonates does not result from their incompetence in terms of myeloid inflammatory response to UVR.


Subject(s)
Macrophages/pathology , Melanoma/pathology , Neoplasms, Radiation-Induced/pathology , Neutrophils/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Age Factors , Animals , Animals, Newborn , Cell Proliferation/radiation effects , Flow Cytometry , Macrophages/radiation effects , Melanoma/epidemiology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Myeloid Cells/radiation effects , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/immunology , Neutrophils/radiation effects , Risk Factors , Skin Neoplasms/epidemiology , Skin Neoplasms/immunology
6.
Methods Mol Biol ; 989: 33-43, 2013.
Article in English | MEDLINE | ID: mdl-23483385

ABSTRACT

The epidermis is a multilayered epithelium consisting of multiple different progenitor cell populations, all of which are important to epidermal function. In order to study these populations, several techniques have been developed that enable specific purification of the different progenitor cell populations. The best characterized stem cell population in the epidermis, and likely the most pluripotent, are the quiescent stem cells in the hair follicle bulge. In this chapter, we provide a method for isolating bulge stem cells from skin of adult mice using fluorescence-activated cell sorting of immunofluorescently labeled keratinocytes. We use the cell surface markers CD34 and α6-integrin for the enrichment of bulge stem cells. This method also contains notes on how to adjust the cytometer settings for a reproducible analysis.


Subject(s)
Flow Cytometry/methods , Stem Cells/cytology , Cell Culture Techniques , Epidermis , Integrin alpha6/metabolism , Keratinocytes/cytology , Skin/cytology , Stem Cells/metabolism
7.
Endocr Relat Cancer ; 20(3): 273-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23404854

ABSTRACT

The intrinsic properties underlying cancer development are extensively studied while the effect of a cancer on the host is often overlooked. Activation of the Hedgehog (Hh) signaling pathway underlies a number of types of common human cancers, yet little is known concerning endocrine signaling in such tumors. Here, we investigated endocrine signaling in a murine model of basal cell carcinoma (BCC) of the skin, the most common cancer. BCCs were generated by the activation of Hh signaling resulting from the specific deletion of the Ptch1 gene in the developing epidermis. Subsequently, a severe growth deficiency was observed in the murine BCC model, and we identified a deficiency of circulating IGF1 (Igf1). We demonstrate that Hh pathway activation in murine BCC induces IGF binding proteins, thereby regulating Igf1 sequestration into the skin and skewing Igf endocrine signaling. Significantly, these results show that Hh-induced tumors can have endocrine effects on normal tissues that in turn can greatly impact the host. This study not only identifies that Igf is important in Hh-associated skin tumors but also exemplifies the need to consider endocrine signaling when interpreting complex in vivo tumor models.


Subject(s)
Carcinoma, Basal Cell/metabolism , Hedgehog Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Skin Neoplasms/metabolism , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Female , Integrases/metabolism , Keratin-14/metabolism , Male , Mice , Mice, Transgenic , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Thymus Gland/metabolism , Thymus Gland/pathology
8.
Cancer Prev Res (Phila) ; 3(10): 1222-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20858761

ABSTRACT

Basal cell carcinoma (BCC) of the skin is the most common form of cancer, with the majority being caused by mutations in the Patched1 (Ptch1) gene, leading to activation of the Hedgehog (Hh) signaling pathway. Hh signaling is implicated in many tumor types; thus, defining the mechanisms by which Ptch1 regulates tissue proliferation is of paramount importance. Here, we show that the key role of Ptch1 in the skin is to limit the size of the epidermal stem/progenitor compartment and allow hair follicle differentiation. Specifically, loss of Ptch1 leads to the promotion of progenitor cell fate by increasing basal cell proliferation and limiting the progression of basal cells into differentiated hair follicle cell types. Our data indicate that BCCs likely result from hair follicle progenitor cells that, due to Hh signal activation, cannot progress through normal hair follicle differentiation. These data confirm the role of Ptch1 as a negative regulator of epidermal progenitor turnover and also show for the first time that Ptch1 plays a role in the differentiation of the hair follicle lineage. In addition, we show that insulin-like growth factor binding protein 2 (Igfbp2) is upregulated in both murine and human BCCs and that blocking Igfbp2 activity reduces the Hh-mediated expansion of epidermal progenitor cells. We propose that Igfbp2 mediates epidermal progenitor cell expansion and therefore represents an epidermal progenitor cell-specific target of Hh signaling that promotes BCC development.


Subject(s)
Carcinoma, Basal Cell/genetics , Cell Transformation, Neoplastic/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Receptors, Cell Surface/genetics , Skin Neoplasms/genetics , Stem Cells/metabolism , Animals , Blotting, Western , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression , Gene Expression Profiling , Hair Follicle/pathology , Hedgehog Proteins/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Insulin-Like Growth Factor Binding Protein 2/metabolism , Mice , Mice, Knockout , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Stem Cells/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...