Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Diabetes ; 73(1): 120-134, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37874683

ABSTRACT

Wound healing is a complex, highly regulated process and is substantially disrupted by diabetes. We show here that human wound healing induces specific epigenetic changes that are exacerbated by diabetes in an animal model. We identified epigenetic changes and gene expression alterations that significantly reduce reepithelialization of skin and mucosal wounds in an in vivo model of diabetes, which were dramatically rescued in vivo by blocking these changes. We demonstrate that high glucose altered FOXO1-matrix metallopeptidase 9 (MMP9) promoter interactions through increased demethylation and reduced methylation of DNA at FOXO1 binding sites and also by promoting permissive histone-3 methylation. Mechanistically, high glucose promotes interaction between FOXO1 and RNA polymerase-II (Pol-II) to produce high expression of MMP9 that limits keratinocyte migration. The negative impact of diabetes on reepithelialization in vivo was blocked by specific DNA demethylase inhibitors in vivo and by blocking permissive histone-3 methylation, which rescues FOXO1-impaired keratinocyte migration. These studies point to novel treatment strategies for delayed wound healing in individuals with diabetes. They also indicate that FOXO1 activity can be altered by diabetes through epigenetic changes that may explain other diabetic complications linked to changes in diabetes-altered FOXO1-DNA interactions. ARTICLE HIGHLIGHTS: FOXO1 expression in keratinocytes is needed for normal wound healing. In contrast, FOXO1 expression interferes with the closure of diabetic wounds. Using matrix metallopeptidase 9 as a model system, we found that high glucose significantly increased FOXO1-matrix metallopeptidase 9 interactions via increased DNA demethylation, reduced DNA methylation, and increased permissive histone-3 methylation in vitro. Inhibitors of DNA demethylation and permissive histone-3 methylation improved the migration of keratinocytes exposed to high glucose in vitro and the closure of diabetic skin and mucosal wounds in vivo. Inhibition of epigenetic enzymes that alter FOXO1-induced gene expression dramatically improves diabetic healing and may apply to other conditions where FOXO1 has a detrimental role in diabetic complications.


Subject(s)
Diabetes Complications , Diabetes Mellitus, Experimental , Animals , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Histones/metabolism , Diabetes Mellitus, Experimental/metabolism , Keratinocytes/metabolism , Diabetes Complications/metabolism , Epigenesis, Genetic , Glucose/metabolism , DNA/metabolism , Re-Epithelialization
3.
Cell Stem Cell ; 28(9): 1582-1596.e6, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34102139

ABSTRACT

Stem cells support lifelong maintenance of adult organs, but their specific roles during injury are poorly understood. Here we demonstrate that Lgr6 marks a regionally restricted population of epidermal stem cells that interact with nerves and specialize in wound re-epithelialization. Diphtheria toxin-mediated ablation of Lgr6 stem cells delays wound healing, and skin denervation phenocopies this effect. Using intravital imaging to capture stem cell dynamics after injury, we show that wound re-epithelialization by Lgr6 stem cells is diminished following loss of nerves. This induces recruitment of other stem cell populations, including hair follicle stem cells, which partially compensate to mediate wound closure. Single-cell lineage tracing and gene expression analysis reveal that the fate of Lgr6 stem cells is shifted toward differentiation following loss of their niche. We conclude that Lgr6 epidermal stem cells are primed for injury response and interact with nerves to regulate their fate.


Subject(s)
Re-Epithelialization , Receptors, G-Protein-Coupled , Epidermal Cells , Hair Follicle , Stem Cells
4.
Cell Stem Cell ; 28(7): 1233-1247.e4, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33984283

ABSTRACT

The functional heterogeneity of resident stem cells that support adult organs is incompletely understood. Here, we directly visualize the corneal limbus in the eyes of live mice and identify discrete stem cell niche compartments. By recording the life cycle of individual stem cells and their progeny, we directly analyze their fates and show that their location within the tissue can predict their differentiation status. Stem cells in the inner limbus undergo mostly symmetric divisions and are required to sustain the population of transient progenitors that support corneal homeostasis. Using in situ photolabeling, we captured their progeny exiting the niche before moving centripetally in unison. The long-implicated slow-cycling stem cells are functionally distinct and display local clonal dynamics during homeostasis but can contribute to corneal regeneration after injury. This study demonstrates how the compartmentalized organization of functionally diverse stem cell populations supports the maintenance and regeneration of an adult organ.


Subject(s)
Epithelium, Corneal , Limbus Corneae , Animals , Cell Differentiation , Cornea , Mice , Stem Cells
6.
J Invest Dermatol ; 141(2): 295-307.e13, 2021 02.
Article in English | MEDLINE | ID: mdl-32649944

ABSTRACT

Squamous cell carcinoma in situ (SCCIS) is a prevalent precancerous lesion that can progress to cutaneous squamous cell carcinoma. Although SCCIS is common, its pathogenesis remains poorly understood. To better understand SCCIS development, we performed laser captured microdissection of human SCCIS and the adjacent epidermis to isolate genomic DNA and RNA for next-generation sequencing. Whole-exome sequencing identified UV-signature mutations in multiple genes, including NOTCH1-3 in the epidermis and SCCIS and oncogenic TP53 mutations in SCCIS. Gene families, including SLFN genes, contained UV/oxidative-signature disruptive epidermal mutations that manifested positive selection in SCCIS. The frequency and distribution of NOTCH and TP53 mutations indicate that NOTCH mutations may precede TP53 mutations. RNA sequencing identified 1,166 differentially expressed genes; the top five enriched gene ontology biological processes included (i) immune response, (ii) epidermal development, (iii) protein phosphorylation, (iv) regulation of catalytic activity, and (v) cytoskeletal regulation. The NEURL1 ubiquitin ligase, which targets Notch ligands for degradation, was upregulated in SCCIS. NEURL1 protein was found to be elevated in SCCIS suggesting that increased levels could represent a mechanism for downregulating Notch during UV-induced carcinogenesis. The data from DNA and RNA sequencing of epidermis and SCCIS provide insights regarding SCCIS formation.


Subject(s)
Carcinoma in Situ/etiology , Carcinoma, Squamous Cell/etiology , Epidermis/radiation effects , Exome , Gene Expression Profiling , Neoplasms, Radiation-Induced/etiology , Skin Neoplasms/etiology , Carcinogenesis/genetics , Carcinoma in Situ/genetics , Carcinoma, Squamous Cell/genetics , Genes, p53 , Humans , Mutation , Neoplasms, Radiation-Induced/genetics , Receptors, Notch/genetics , Sequence Analysis, RNA , Skin Neoplasms/genetics , Ultraviolet Rays
7.
Cell Mol Gastroenterol Hepatol ; 10(4): 868-880.e1, 2020.
Article in English | MEDLINE | ID: mdl-32376419

ABSTRACT

BACKGROUND & AIMS: Female sex is associated with lower incidence and improved clinical outcomes for most cancer types including pancreatic ductal adenocarcinoma (PDAC). The mechanistic basis for this sex difference is unknown. We hypothesized that estrogen signaling may be responsible, despite the fact that PDAC lacks classic nuclear estrogen receptors. METHODS: Here we used murine syngeneic tumor models and human xenografts to determine that signaling through the nonclassic estrogen receptor G protein-coupled estrogen receptor (GPER) on tumor cells inhibits PDAC. RESULTS: Activation of GPER with the specific, small molecule, synthetic agonist G-1 inhibited PDAC proliferation, depleted c-Myc and programmed death ligand 1 (PD-L1), and increased tumor cell immunogenicity. Systemically administered G-1 was well-tolerated in PDAC bearing mice, induced tumor regression, significantly prolonged survival, and markedly increased the efficacy of PD-1 targeted immune therapy. We detected GPER protein in a majority of spontaneous human PDAC tumors, independent of tumor stage. CONCLUSIONS: These data, coupled with the wide tissue distribution of GPER and our previous work showing that G-1 inhibits melanoma, suggest that GPER agonists may be useful against a range of cancers that are not classically considered sex hormone responsive and that arise in tissues outside of the reproductive system.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Receptors, G-Protein-Coupled/agonists , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
8.
Exp Dermatol ; 29(1): 29-38, 2020 01.
Article in English | MEDLINE | ID: mdl-31519066

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer and is associated with cumulative UV exposure. Studies have shown that prolonged voriconazole use promotes cSCC formation; however, the biological mechanisms responsible for the increased incidence remain unclear. Here, we show that voriconazole directly increases oxidative stress in human keratinocytes and promotes UV-induced DNA damage as determined by comet assay, 8-oxoguanine immunofluorescence and mass spectrometry. Voriconazole treatment of human keratinocytes potentiates UV-induced apoptosis and activation of the p38 MAP kinase and 53BP1 UV stress response pathways. The p38 MAP kinase activation promoted by voriconazole exposure can be mitigated by pretreating keratinocytes with N-acetylcysteine. Voriconazole increases oxidative stress in keratinocytes by directly inhibiting catalase leading to lower intracellular NADPH levels and the triazole moieties in voriconazole are critical for inhibiting catalase. Furthermore, voriconazole is shown to promote UV-induced dysplasia in an in vivo model. Together, these data demonstrate that voriconazole potentiates oxidative stress in UV-irradiated keratinocytes through catalase inhibition. Use of antioxidants may mitigate the pro-oncogenic effects of voriconazole.


Subject(s)
Antifungal Agents/pharmacology , DNA Damage/drug effects , Oxidative Stress/drug effects , Ultraviolet Rays/adverse effects , Voriconazole/pharmacology , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , Catalase/antagonists & inhibitors , Cell Proliferation/drug effects , Cells, Cultured , DNA Damage/radiation effects , Humans , Keratinocytes/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mice , Primary Cell Culture , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin/radiation effects , Terbinafine/pharmacology , Tumor Suppressor p53-Binding Protein 1/metabolism
9.
Exp Dermatol ; 28(5): 609-613, 2019 05.
Article in English | MEDLINE | ID: mdl-30762245

ABSTRACT

Actinic keratoses (AKs) and squamous cell carcinoma in situ (SCCIS) are precursor lesions for cutaneous squamous cell carcinoma (cSCC), the second most common form of cancer. Current topical therapies for AKs and SCCIS promote skin inflammation to eradicate lesions and do not directly target the biological mechanisms driving growth. We hypothesized that topical small molecule inhibitors targeting kinases promoting keratinocyte growth in AKs and SCCIS could induce regression of these lesions with less inflammation. To test this hypothesis, we determined the efficacy of topical dasatinib, 5-fluorouracil and BEZ-235 in inducing regression of cSCCs in the K14-Fyn Y528 transgenic mouse model. Topical dasatinib induced regression of cSCC with less inflammation, no ulceration and no mortality compared to 5-fluorouracil. Topical BEZ-235 induced cSCC regression similar to dasatinib without erythema or ulceration. These data indicate that topical small molecule kinase inhibitors targeting drivers of AK/SCCIS/cSCC growth represent a promising therapeutic approach to treat these common skin lesions.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Keratosis, Actinic/drug therapy , Protein Kinase Inhibitors/administration & dosage , Skin Neoplasms/drug therapy , Administration, Topical , Animals , Dasatinib/administration & dosage , Fluorouracil/administration & dosage , Humans , Imidazoles/administration & dosage , Inflammation , Keratinocytes/pathology , Mice , Mice, Transgenic , Quinolines/administration & dosage , Treatment Outcome
10.
J Cutan Pathol ; 46(5): 310-316, 2019 May.
Article in English | MEDLINE | ID: mdl-30666677

ABSTRACT

BACKGROUND: Accurate classification of spitzoid melanocytic lesions is difficult due to overlapping clinical and histopathologic features between Spitz nevi, atypical Spitz tumors (ASTs), and spitzoid melanomas. Expression of p16 (CDKN2A) has been used as a marker of spitzoid lesions. However, its expression may be variable. p15 is a tumor suppressor encoded by CDKN2B, loss of which has been recently shown to promote transition from nevus to melanoma. We sought to determine whether p15 is a useful immunohistochemical marker to distinguish Spitz nevi from spitzoid melanomas and to compare p15 and p16 staining in this population. METHODS: Immunohistochemistry for p15 and p16 was performed on Spitz nevi (n = 19), ASTs (n = 41), and spitzoid melanomas (n = 17). Immunoexpression was categorized by a four-tiered system: 0 (negative), 1+ (weak), 2+ (moderate), 3+ (strong). RESULTS: 3+/strong p15 staining was observed in 68.4% of Spitz nevi, 34.2% of ASTs, and 17.7% of spitzoid melanomas. By contrast, we observed 3+ p16 staining in roughly equivalent percentages of Spitz nevi (57.9%), ASTs (56.1%), and spitzoid melanomas (58.8%). CONCLUSION: These data illustrate that p15 may be more useful than p16 as a biomarker to help distinguish benign from malignant spitzoid lesions.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Melanoma , Nevus, Epithelioid and Spindle Cell , Skin Neoplasms , Female , Humans , Immunohistochemistry , Male , Melanoma/metabolism , Melanoma/pathology , Nevus, Epithelioid and Spindle Cell/metabolism , Nevus, Epithelioid and Spindle Cell/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
11.
Elife ; 72018 01 16.
Article in English | MEDLINE | ID: mdl-29336307

ABSTRACT

Female sex and history of prior pregnancies are associated with favorable melanoma outcomes. Here, we show that much of the melanoma protective effect likely results from estrogen signaling through the G protein-coupled estrogen receptor (GPER) on melanocytes. Selective GPER activation in primary melanocytes and melanoma cells induced long-term changes that maintained a more differentiated cell state as defined by increased expression of well-established melanocyte differentiation antigens, increased pigment production, decreased proliferative capacity, and decreased expression of the oncodriver and stem cell marker c-Myc. GPER signaling also rendered melanoma cells more vulnerable to immunotherapy. Systemically delivered GPER agonist was well tolerated, and cooperated with immune checkpoint blockade in melanoma-bearing mice to dramatically extend survival, with up to half of mice clearing their tumor. Complete responses were associated with immune memory that protected against tumor rechallenge. GPER may be a useful, pharmacologically accessible target for melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Receptors, G-Protein-Coupled/agonists , Signal Transduction , Animals , Disease Models, Animal , Humans , Mice , Pigments, Biological , Receptors, Estrogen , Survival Analysis , Treatment Outcome , Tumor Cells, Cultured
12.
Am J Pathol ; 186(12): 3094-3099, 2016 12.
Article in English | MEDLINE | ID: mdl-27855847

ABSTRACT

Most melanomas are driven by BRAF(V600E)-activating mutations, while nevi harboring the same mutations have growth arrest. Although decreased p16 expression has been associated with melanoma formation, in recent work, p15 represented a primary effector of oncogene-induced senescence in nevomelanocytes that was diminished in melanomas. This study determined whether decreased p15 levels represent a general biomarker for the transition from nevus to melanoma. We performed p15 and p16 IHC analyses on a random series of nevi and melanomas. Staining was evaluated and graded for percentage and intensity to determine the H score. For real-time quantitative RT-PCR analysis of p15, RNA was extracted from FFPE sections from 14 nevus and melanoma samples via macrodissection. A two-sided t-test was used to evaluate between-group differences in mean H scores and qΔCt values. p15 Expression was significantly increased in melanocytic nevi compared with melanomas (mean H scores, 254.8 versus 132.3; P < 0.001). On p15 staining, the H score differential was greater than that with p16 staining [122.5 (P < 0.001) and 64.8 (P = 0.055), respectively]. Real-time quantitative RT-PCR analysis revealed a lower mean qΔCt value in melanomas, consistent with lower p15 expression (P = 0.018). Together, these data support the hypothesis that decreased p15 expression is a robust biomarker for distinguishing nevus from melanoma.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Melanoma/metabolism , Nevus, Pigmented/metabolism , Nevus/metabolism , Skin Neoplasms/metabolism , Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Diagnosis, Differential , Humans , Melanocytes/metabolism , Melanoma/pathology , Mutation , Nevus/pathology , Nevus, Pigmented/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology
13.
Cancer Discov ; 5(10): 1072-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26183406

ABSTRACT

UNLABELLED: Deletion of the entire CDKN2B-CDKN2A gene cluster is among the most common genetic events in cancer. The tumor-promoting effects are generally attributed to loss of CDKN2A-encoded p16 and p14ARF tumor suppressors. The degree to which the associated CDKN2B-encoded p15 loss contributes to human tumorigenesis is unclear. Here, we show that CDKN2B is highly upregulated in benign melanocytic nevi, contributes to maintaining nevus melanocytes in a growth-arrested premalignant state, and is commonly lost in melanoma. Using primary melanocytes isolated directly from freshly excised human nevi naturally expressing the common BRAF(V600E)-activating mutation, nevi progressing to melanoma, and normal melanocytes engineered to inducibly express BRAF(V600E), we show that BRAF activation results in reversible, TGFß-dependent, p15 induction that halts proliferation. Furthermore, we engineer human skin grafts containing nevus-derived melanocytes to establish a new, architecturally faithful, in vivo melanoma model, and demonstrate that p15 loss promotes the transition from benign nevus to melanoma. SIGNIFICANCE: Although BRAF(V600E) mutations cause melanocytes to initially proliferate into benign moles, mechanisms responsible for their eventual growth arrest are unknown. Using melanocytes from human moles, we show that BRAF activation leads to a CDKN2B induction that is critical for restraining BRAF oncogenic effects, and when lost, contributes to melanoma.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/deficiency , Cyclin-Dependent Kinase Inhibitor p15/genetics , Melanoma/genetics , Melanoma/pathology , Nevus/genetics , Nevus/pathology , Animals , Cell Cycle Checkpoints/genetics , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA Mutational Analysis , Disease Models, Animal , Disease Progression , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunohistochemistry , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Mice , Mutation , Nevus/metabolism , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction , Transcriptional Activation , Transforming Growth Factor beta/metabolism
15.
Sci Transl Med ; 7(275): 275ra22, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25696001

ABSTRACT

Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv-based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII(+) glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).


Subject(s)
Brain Neoplasms/therapy , ErbB Receptors/immunology , Glioblastoma/therapy , Immunotherapy , Receptors, Antigen, T-Cell/immunology , Animals , Disease Models, Animal , Heterografts , Humans , Mice
16.
PLoS One ; 9(6): e100800, 2014.
Article in English | MEDLINE | ID: mdl-24959865

ABSTRACT

The skin serves multiple functions that are critical for life. The protection from pathogens is achieved by a complicated interaction between aggressive effectors and controlling functions that limit damage. Inhomogeneous radiation with limited penetration is used in certain types of therapeutics and is experienced with exposure to solar particle events outside the protection of the Earth's magnetic field. This study explores the effect of ionizing radiation on skin immune function. We demonstrate that radiation, both homogeneous and inhomogeneous, induces inflammation with resultant specific loss of regulatory T cells from the skin. This results in a hyper-responsive state with increased delayed type hypersensitivity in vivo and CD4+ T cell proliferation in vitro. The effects of inhomogeneous radiation to the skin of astronauts or as part of a therapeutic approach could result in an unexpected enhancement in skin immune function. The effects of this need to be considered in the design of radiation therapy protocols and in the development of countermeasures for extended space travel.


Subject(s)
Radiation Injuries, Experimental/immunology , Skin/radiation effects , T-Lymphocytes, Regulatory/radiation effects , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/radiation effects , Female , Male , Mice, Inbred ICR , Radiation, Ionizing , Swine
17.
Hum Mol Genet ; 23(15): 4001-14, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24634144

ABSTRACT

Understanding the influence of gene expression on the molecular mechanisms underpinning human phenotypic diversity is fundamental to being able to predict health outcomes and treat disease. We have carried out whole transcriptome expression analysis on a series of eight normal human postmortem eyes by RNA sequencing. Here we present data showing that ∼80% of the transcriptome is expressed in the posterior layers of the eye and that there is significant differential expression not only between the layers of the posterior part of the eye but also between locations of a tissue layer. These differences in expression also extend to alternative splicing and splicing factors. Differentially expressed genes are enriched for genes associated with psychiatric, immune and cardiovascular disorders. Enrichment categories for gene ontology included ion transport, synaptic transmission and visual and sensory perception. Lastly, allele-specific expression was found to be significant for CFH, C3 and CFB, which are known risk genes for age-related macular degeneration. These expression differences should be useful in determining the underlying biology of associations with common diseases of the human retina, retinal pigment epithelium and choroid and in guiding the analysis of the genomic regions involved in the control of normal gene expression.


Subject(s)
Choroid/metabolism , Eye Proteins/genetics , Retinal Pigment Epithelium/metabolism , Transcriptome , Aged , Aged, 80 and over , Autopsy , Complement C3/genetics , Complement Factor B/genetics , Complement Factor H/genetics , Gene Expression Profiling , Genome-Wide Association Study , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Metabolic Networks and Pathways/genetics , Middle Aged , Molecular Sequence Annotation , Risk Factors
18.
J Invest Dermatol ; 134(1): 229-236, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23792457

ABSTRACT

Sézary syndrome (SS) cells express cell surface molecules also found on normal activated CD4 T cells. In an effort to find a more specific surface marker for malignant SS cells, a microarray analysis of gene expression was performed. Results showed significantly increased levels of mRNA for CD164, a sialomucin found on human CD34+ hematopoietic stem cells, and FCRL3, a molecule present on a subset of human natural T regulatory cells. Both markers were increased in CD4 T cells from SS patients compared with healthy donors (HD). Flow cytometry studies confirmed the increased expression of CD164 and FCRL3 primarily on CD4+CD26- T cells of SS patients. Importantly, a statistically significant correlation was found between an elevated percentage of CD4+CD164+ T cells and an elevated percentage of CD4+CD26- T cells in all tested SS patients but not in patients with mycosis fungoides and atopic dermatitis or HD. FCRL3 expression was significantly increased only in patients with high tumor burden. CD4+CD164+ cells displayed cerebriform morphology and their loss correlated with clinical improvement in treated patients. Our results suggest that CD164 can serve as a marker for diagnosis and for monitoring progression of cutaneous T-cell lymphoma (CTCL)/SS and that FCRL3 expression correlates with a high circulating tumor burden.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endolyn/immunology , Neoplastic Cells, Circulating/immunology , Receptors, Immunologic/immunology , Sezary Syndrome/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Shape/immunology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/immunology , Dipeptidyl Peptidase 4/metabolism , Disease Progression , Endolyn/genetics , Endolyn/metabolism , Flow Cytometry , Humans , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Neoplastic Cells, Circulating/metabolism , Receptors, Immunologic/metabolism , Transcriptome
20.
Arch Dermatol ; 148(2): 219-23, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22351821

ABSTRACT

BACKGROUND: Viral-associated trichodysplasia of immunosuppression is a rare cutaneous eruption that is characterized by follicularly based shiny papules and alopecia with characteristic histopathologic findings of abnormally anagen follicules with excessive inner root sheath differentiation. Prior reports have described the histopathologic characteristics on vertical sections; however, to our knowledge, immunohistochemical analysis of polyomavirus proteins has not been previously performed. OBSERVATIONS: We discuss the thorough diagnostic evaluation and therapy of an unusual case of viral-associated trichodysplasia due to a newly described human polyomavirus that occurred in a patient with posttreatment chronic lymphocytic leukemia and an abnormal white blood cell count. Unique to our study is the immunohistochemical staining for the polyomavirus middle T antigen, which demonstrated positive staining of cellular inclusions within keratinocytes that compose the inner root sheath. Further evaluation with scanning electron microscopy and polymerase chain reaction analysis of viral DNA confirmed the presence of the virus. Treatment with topical cidofovir resulted in dramatic clinical improvement and hair regrowth. CONCLUSIONS: Several tools, including immunohistochemical staining for the polyomavirus middle T antigen, can be used to identify the pathogenic virus associated with viral-associated trichodysplasia. This case highlights the utility of multiple diagnostic modalities and a robust response to a topical therapeutic agent, cidofovir.


Subject(s)
Antiviral Agents/therapeutic use , Cytosine/analogs & derivatives , DNA, Viral/analysis , Hair Diseases/drug therapy , Organophosphonates/therapeutic use , Polyomavirus Infections/drug therapy , Cidofovir , Cytosine/therapeutic use , Female , Hair Diseases/pathology , Hair Diseases/virology , Hair Follicle/chemistry , Hair Follicle/pathology , Hair Follicle/virology , Humans , Middle Aged , Polyomavirus , Polyomavirus Infections/metabolism , Polyomavirus Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL