Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 8429, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38600101

ABSTRACT

Vulvar lichen sclerosus (VLS) is a chronic and progressive dermatologic condition that can cause physical dysfunction, disfigurement, and impaired quality of life. However, the etiology of VLS remains unknown. The vulvar skin, intestinal and vaginal microbiomes have been postulated to play important roles in the pathogenesis of this disease. The aim of this study was to compare the compositional characteristics of the vulvar skin, vagina, and gut microbiota between perimenopausal or postmenopausal VLS patients and healthy controls. The study involved six perimenopausal or postmenopausal VLS patients which were based on characteristic clinical manifestations and histologic confirmation and five healthy controls. The pruritus severity of each patient was evaluated using the NRS scale, and the dermatology-specific health-related quality of life was assessed using the Skindex-16. Metagenomic sequencing was performed, and the results were analyzed for alpha and beta diversity. LEfSe analysis were used to investigate the microbial alterations in vulvar skin, gut and vagina. KEGG databases were used to analyze differences in functional abundance. The study found significant differences in alpha diversity between the two groups in stool and vaginal samples (P < 0.05). Patients with VLS had a higher abundance of Enterobacter cloacae, Flavobacterium_branchiophilum, Mediterranea_sp._An20, Parabacteroides_johnsoniiand Streptococcus_bovimastitidis on the vulvar skin, while Corynebacterium_sp._zg-913 was less abundant compared to the control group. The relative abundance of Sphingomonas_sp._SCN_67_18, Sphingobium_sp._Ant17, and Pontibacter_sp_BT213 was significantly higher in the gut samples of patients with VLS.Paenibacillus_popilliae,Gemella_asaccharolytica, and Coriobacteriales_bacterium_DNF00809 compared to the control group. Additionally, the vaginal samples of patients with VLS exhibited a significantly lower relative abundance of Bacteroidales_bacterium_43_8, Bacteroides_sp._CAG:20, Blautia_sp._AM28-10, Fibrobacter_sp._UWB16, Lachnospiraceae_bacterium_AM25-39, Holdemania_filiformis, Lachnospiraceae_bacterium_GAM79, and Tolumonas_sp. Additionally, the butyrate-producing bacterium SS3/4 showed a significant difference compared to the controls. The study found a negative relationship between Sphingobium_sp._Ant17 in stool and Skindex-16 (P < 0.05), while Mediterranea_sp._An20 had a positive correlation with Skindex-16 (P < 0.05) in the skin. Additionally, our functional analysis revealed alterations in Aminoacyl_tRNA_biosynthesis, Glutathione_metabolism, the pentose phosphate pathway, and Alanine__aspartate_and_glutamate_metabolism in the VLS patient group. The study suggests that perimenopausal or postmenopausal patients with VLS have a modified microbiome in the vulvar skin, gut, and vagina. This modification is linked to abnormal energy metabolism, increased oxidative stress, and abnormal amino acid metabolism.


Subject(s)
Microbiota , Vulvar Lichen Sclerosus , Female , Humans , Vulvar Lichen Sclerosus/pathology , Postmenopause , Perimenopause , Quality of Life , Arrhythmias, Cardiac , Vagina/pathology
2.
Clin Cosmet Investig Dermatol ; 16: 3521-3525, 2023.
Article in English | MEDLINE | ID: mdl-38089016

ABSTRACT

Atopic dermatitis and psoriasis are both common chronic inflammatory skin conditions that can significantly affect the quality of life for individuals affected by them. With the growing use of biologic agents, there have been remarkable clinical advancements in the treatment of these diseases. Interestingly, during biologic therapy for either condition, a phenomenon has emerged where treatment can paradoxically induce a transition to the phenotype of the other condition.We present two cases of immune drift phenomena caused by biologic agents for treating psoriasis and atopic dermatitis.The first one is a case of psoriasis lesion that developed in an old patient with AD who was treated with dupilumab for two months. The second one is a case of eczematoid lesion that developed in an adult patient with ankylosing spondylitis who was treated with Secukinumab for 1 year.

3.
Exp Dermatol ; 31(3): 320-329, 2022 03.
Article in English | MEDLINE | ID: mdl-34623712

ABSTRACT

Abnormal activation of fibroblasts plays a crucial role in keloid development. However, the mechanism of fibroblast activation remains to be determined. YAP/TAZ are key molecules in the Hippo signalling pathway that promote cell proliferation and inhibit apoptosis. Here, we show that keloid fibroblasts have higher levels of YAP/TAZ mRNA and proteins on primary culture. Targeted knockdown of endogenous YAP or TAZ significantly inhibited cell proliferation, reduced cell migration, induced cell apoptosis and down-regulated collagen1a1 production by keloid fibroblasts. Moreover, we demonstrate that verteporfin, an inhibitor of YAP/TAZ, has similar but stronger inhibitory effects on fibroblasts compared to YAP/TAZ knockdown. Our study provides evidence that YAP/TAZ may be involved in the pathogenesis of keloids. Targeted inhibition of YAP/TAZ could change the biological behaviours of fibroblasts and can potentially be used as therapy for keloids.


Subject(s)
Keloid , Fibroblasts/metabolism , Humans , Keloid/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Verteporfin/metabolism , Verteporfin/pharmacology
4.
J Invest Dermatol ; 142(2): 390-401, 2022 02.
Article in English | MEDLINE | ID: mdl-34293351

ABSTRACT

Psoriasis is a chronic immune-mediated disease characterized by excessive proliferation of epidermal keratinocytes and increased immune cell infiltration to the skin. Although it is well-known that psoriasis pathogenesis is driven by aberrant production of proinflammatory cytokines, the mechanisms underlying the imbalance between proinflammatory and anti-inflammatory cytokine expression are incompletely understood. In this study, we report that the transcriptional coregulators CtBP1 and 2 can transactivate a common set of proinflammatory genes both in the skin of imiquimod-induced mouse psoriasis model and in human keratinocytes and macrophages stimulated by imiquimod. We find that mice overexpressing CtBP1 in epidermal keratinocytes display severe skin inflammation phenotypes with increased expression of T helper type 1 and T helper type 17 cytokines. We also find that the expression of CtBPs and CtBP-target genes is elevated both in human psoriatic lesions and in the mouse imiquimod psoriasis model. Moreover, we were able to show that topical treatment with a peptidic inhibitor of CtBP effectively suppresses the CtBP-regulated proinflammatory gene expression and thus attenuates psoriatic inflammation in the imiquimod mouse model. Together, our findings suggest to our knowledge previously unreported strategies for therapeutic modulation of the immune response in inflammatory skin diseases.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Psoriasis/drug therapy , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , HaCaT Cells , Humans , Imiquimod/immunology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology , Mice , Mice, Transgenic , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
5.
Lipids Health Dis ; 19(1): 171, 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32677954

ABSTRACT

BACKGROUND: Senile pruritus is common, yet its etiology remains unknown. Aging-associated skin barrier defects and skin surface lipid (SSL) alterations have been postulated to play important roles in its occurrence. In the present study, the lipidomic profiles of SSLs in elderly patients were examined to better understand the potential causes of senile pruritus. METHODS: Transepidermal water loss (TEWL) was evaluated to assess the skin barrier function. The Ameliorated Kawashima Itch Scale score was used to measure the pruritus severity. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) and multivariate data analysis were employed to investigate SSL alterations. RESULTS: The results showed that senile pruritus patients had higher TEWL values than control subjects (13.13 ± 4.28 versus 6.71 ± 2.45, p < 0.01). LC-MS/MS revealed significant differences in the lipidomic profiles and identified 81 species of SSLs that differed between the two groups. Compared with control subjects, senile pruritus patients had increased levels of ceramides (Cers), diacylglycerols, fatty acids, phosphatidylcholines, phosphatidylethanolamines, phytosphingosines, sphingosines, diacylceryl-3-O-carboxyhydroxymethylcholine, diacylglyceryl trimethylhomoserine, and unsaturated free fatty acids, but decreased levels of triacylglycerol. Cer-EOS, Cer-NDS, and Cer-NS were positively correlated with TEWL value (p < 0.05). Pruritus severity score was positively correlated with sphingomyelin, Cer-NP, Cer-AS, Cer-NDS, and Cer-NS, but negatively correlated with Cer-BS, Cer-EODS, Cer-EOS, and Cer-AP. CONCLUSIONS: The present study indicated that patients with senile pruritus have impaired skin barrier function and altered SSL composition. Certain SSL species identified in this study may be potential targets for future studies on the pathogenesis of senile pruritus. TRIAL REGISTRATION: Peking University International Hospital (Number: YN2018QN04 ; date: January 2019).


Subject(s)
Lipidomics/methods , Pruritus/metabolism , Skin/metabolism , Aged , Ceramides/metabolism , Chromatography, Liquid , Diglycerides/metabolism , Female , Humans , Male , Middle Aged , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tandem Mass Spectrometry
6.
World J Surg Oncol ; 18(1): 56, 2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32183822

ABSTRACT

BACKGROUND: Numerous literatures have demonstrated that circular RNAs (circRNAs) are involved in multiple types of tumors. However, the effects of circRNAs in melanoma are not very clear. In this study, we aimed to investigate the roles and mechanisms of circ-FOXM1 in melanoma. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine the expression of circ-FOXM1, microRNA-143-3p (miR-143-3p), and Flotillin 2 (FLOT2) mRNA. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, flow cytometry analysis, and transwell assay were employed to test cell proliferation, apoptosis, and invasion, respectively. The glucose consumption and lactate production were examined by specific kits. Western blot assay was utilized for the detection of hexokinase2 (HK2), pyruvate kinase isozyme type M2 (PKM2), and FLOT2. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were employed to verify the targeting association between miR-143-3p and circ-FOXM1 or FLOT2. A murine xenograft model was established to explore the effect of circ-FOXM1 in vivo. RESULTS: Circ-FOXM1 was elevated and miR-143-3p was reduced in melanoma tissues and cells. Circ-FOXM1 deficiency impeded cell proliferation, invasion, and glycolysis and facilitated cell apoptosis in melanoma in vitro and tumorigenesis in vivo. Circ-FOXM1 acted as a sponge of miR-143-3p and the impacts of circ-FOXM1 silencing on cell proliferation, apoptosis, invasion, and glycolysis were overturned by miR-143-3p deletion. Moreover, FLOT2 was a target gene of miR-143-3p and FLOT2 overexpression rescued the inhibitory effect of miR-143-3p on melanoma progression. CONCLUSION: Circ-FOXM1 facilitated the development of melanoma by upregulating FLOT2 through miR-143-3p.


Subject(s)
Melanoma/pathology , Membrane Proteins/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Animals , Apoptosis , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Melanoma/genetics , Melanoma/metabolism , Mice
7.
Clin Cancer Res ; 26(12): 3058-3070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32139402

ABSTRACT

PURPOSE: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). EXPERIMENTAL DESIGN: We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed. RESULTS: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage-associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. CONCLUSIONS: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Disease Models, Animal , Head and Neck Neoplasms/radiotherapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Smad4 Protein/deficiency , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Apoptosis , Cell Proliferation , Cetuximab/administration & dosage , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, Nude , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prognosis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Invest Dermatol ; 139(4): 940-950, 2019 04.
Article in English | MEDLINE | ID: mdl-30423327

ABSTRACT

We assessed the roles of Smad7 in skin inflammation and wound healing using genetic and pharmacological approaches. In K5.TGFß1/K5.Smad7 bigenic (double transgenic) mice, Smad7 transgene expression reversed transforming growth factor (TGF)-ß1 transgene-induced inflammation, fibrosis, and subsequent epidermal hyperplasia and molecularly abolished TGF-ß and NF-κB activation. Next, we produced recombinant human Smad7 protein with a Tat-tag (Tat-Smad7) that rapidly enters cells. Subcutaneous injection of Tat-Smad7 attenuated infiltration of F4/80+ and CD11b+ leukocytes and α-smooth muscle actin+ fibroblasts before attenuating epidermal hyperplasia in K5.TGFß1 skin. Furthermore, topically applied Tat-Smad7 on K5.TGFß1 skin wounds accelerated wound closure, with improved re-epithelialization and reductions in inflammation and fibrotic response. A short treatment with Tat-Smad7 was also sufficient to reduce TGF-ß and NF-κB signaling in K5.TGFß1 skin and wounds. Relevant to the clinic, we found that human diabetic wounds had elevated TGF-ß and NF-κB signaling compared with normal skin. To assess the oncogenic risk of a potential Smad7-based therapy, we exposed K5.Smad7 skin to chemical carcinogenesis and found reduced myeloid leukocyte infiltration in tumors but not accelerated carcinogenesis compared with wild-type littermates. Our study suggests the feasibility of using exogenous Smad7 below an oncogenic level to alleviate skin inflammation and wound healing defects associated with excessive activation of TGF-ß and NF-κB.


Subject(s)
Gene Expression Regulation, Neoplastic , Inflammation/genetics , Neoplasms, Experimental , Skin Neoplasms/genetics , Smad7 Protein/genetics , Transforming Growth Factor beta/metabolism , Wound Healing/genetics , Animals , Carcinogenesis , Guinea Pigs , Humans , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Transgenic , Phenotype , RNA, Neoplasm/genetics , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Smad7 Protein/biosynthesis
9.
Int J Biol Sci ; 14(10): 1163-1174, 2018.
Article in English | MEDLINE | ID: mdl-30123066

ABSTRACT

The endogenous small GTPase, Rac1, plays a critical role during normal skin wound healing. It remains to be determined whether endogenous Rac1 can be appropriately activated in chronic wounds; if not, whether exogenous Rac1 has therapeutic effects on wound healing. Here we show that Rac1 protein levels were lower in wounds of db/db diabetic mice than wounds in wild type mice during the healing process. To assess the therapeutic potential of exogenous Rac1 in wound healing, we produced a Tat-Rac1 fusion protein that enters into cells through protein transduction. Tat-Rac1 increased proliferation and migration of keratinocytes and dermal fibroblasts in vitro. Topical application of Tat-Rac1 accelerated cutaneous wound closure in vivo in db/db mice as well as wild type mice. Further analyses revealed that Tat-Rac1 had faster re-epithelialization, higher keratinocyte proliferation and migration without an earlier onset of myofibroblast activation than vehicle treated wounds. Tat-Rac1 also reduced inflammation in wounds. Our findings revealed the failure of diabetic wounds to elevate Rac1 expression and suggested a therapeutic strategy utilizing a Rac1-based biologic to compensate for this defect thereby promoting wound healing.


Subject(s)
Cell Movement/physiology , Wound Healing/physiology , rac1 GTP-Binding Protein/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Blotting, Western , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Skin/cytology , Wound Healing/genetics , rac1 GTP-Binding Protein/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics
10.
Int J Biol Sci ; 13(3): 319-326, 2017.
Article in English | MEDLINE | ID: mdl-28367096

ABSTRACT

Keloid is a common and refractory disease characterized by abnormal fibroblast proliferation and excessive deposition of extracellular matrix components. Hypocrellin B (HB) is a natural perylene quinone photosensitizer. In this experiment, we studied the effects of photodynamic therapy (PDT) using yellow light from light-emitting diode (LED) combined with HB on keloid fibroblasts (KFB) in vitro. Our results showed that HB-LED PDT treatment induced significant KFB apoptosis and decreased KFB cell viability. HB-LED PDT treatment lead to significant BAX upregulation and BCL-2 downregulation in KFB cells, which led to elevation of intracellular free Ca2+ and activation of caspase-3. Our data provides preliminary evidence for the potential of HB-LED PDT as a therapeutic strategy for keloid.


Subject(s)
Apoptosis/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Keloid , Light , Perylene/analogs & derivatives , Photochemotherapy/methods , Quinones/pharmacology , Perylene/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
Protein Cell ; 6(3): 160-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25566830

ABSTRACT

Oral mucositis, a severe oral ulceration, is a common toxic effect of radio- or chemoradio-therapy and a limiting factor to using the maximum dose of radiation for effective cancer treatment. Among cancer patients, at least 40% and up to 70%, of individuals treated with standard chemotherapy regimens or upper-body radiation, develop oral mucositis. To date, there is no FDA approved drug to treat oral mucositis in cancer patients. The key challenges for oral mucositis treatment are to repair and protect ulcerated oral mucosa without promoting cancer cell growth. Oral mucositis is the result of complex, multifaceted pathobiology, involving a series of signaling pathways and a chain of interactions between the epithelium and submucosa. Among those pathways and interactions, the activation of nuclear factor-kappa B (NF-κB) is critical to the inflammation process of oral mucositis. We recently found that activation of TGFß (transforming growth factor ß) signaling is associated with the development of oral mucositis. Smad7, the negative regulator of TGFß signaling, inhibits both NF-κB and TGFß activation and thus plays a pivotal role in the prevention and treatment of oral mucositis by attenuating growth inhibition, apoptosis, and inflammation while promoting epithelial migration. The major objective of this review is to evaluate the known functions of Smad7, with a particular focus on its molecular mechanisms and its function in blocking multiple pathological processes in oral mucositis.


Subject(s)
Mouth Diseases/metabolism , Mouth Diseases/prevention & control , Smad7 Protein/metabolism , Stomatitis/metabolism , Stomatitis/prevention & control , Animals , Humans , Mouth Diseases/pathology , Stomatitis/pathology
12.
J Invest Dermatol ; 134(5): 1323-1331, 2014 May.
Article in English | MEDLINE | ID: mdl-24280726

ABSTRACT

Carboxyl-terminal-binding protein-1 (CtBP1) is a transcriptional corepressor with multiple in vitro targets, but its in vivo functions are largely unknown. We generated keratinocyte-specific CtBP1 transgenic mice with a keratin-5 promoter (K5.CtBP1) to probe the pathological roles of CtBP1. At transgene expression levels comparable to endogenous CtBP1 in acute skin wounds, the K5.CtBP1 epidermis displayed hyperproliferation, loss of E-cadherin, and failed terminal differentiation. Known CtBP1 target genes associated with these processes, e.g., p21, Brca1, and E-cadherin, were downregulated in K5.CtBP1 skin. Surprisingly, K5.CtBP1 pups also exhibited a hair loss phenotype. We found that expression of the Distal-less 3 (Dlx3), a critical regulator of hair follicle differentiation and cycling, was decreased in K5.CtBP1 mice. Molecular studies revealed that CtBP1 directly suppressed Dlx3 transcription. Consistently, K5.CtBP1 mice displayed abnormal hair follicles with decreased expression of Dlx3 downstream targets Gata3, Hoxc13, and hair keratins. In summary, this CtBP1 transgenic model provides in vivo evidence for certain CtBP1 functions predicted from in vitro studies, reveals--to our knowledge--previously unreported functions and transcriptional activities of CtBP1 in the context of epithelial-mesenchymal interplay, and suggests that CtBP1 has a pathogenic role in hair follicle morphogenesis and differentiation.


Subject(s)
Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hair Follicle/physiology , Homeostasis/genetics , Keratinocytes/physiology , Alopecia/genetics , Alopecia/metabolism , Alopecia/pathology , Animals , Cadherins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Epidermal Cells , Epidermis/physiology , Gene Expression/physiology , Hair Follicle/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Keratin-5/genetics , Keratinocytes/cytology , Mice , Mice, Transgenic , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/physiology
13.
J Clin Invest ; 123(10): 4390-404, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999427

ABSTRACT

Squamous cell carcinomas (SCCs) originate in stratified epithelia, with a small subset becoming metastatic. Epithelial stem cells are targets for driver mutations that give rise to SCCs, but it is unknown whether they contribute to oncogenic multipotency and metastasis. We developed a mouse model of SCC by targeting two frequent genetic mutations in human SCCs, oncogene Kras(G12D) activation and Smad4 deletion, to mouse keratin 15-expressing (K15+) stem cells. We show that transgenic mice developed multilineage tumors, including metastatic SCCs. Among cancer stem cell-enriched (CSC-enriched) populations, those with increased side population (SP) cells correlated with epithelial-mesenchymal transition (EMT) and lung metastasis. We show that microRNA-9 (miR-9) contributed to SP expansion and metastasis, and miR-9 inhibition reduced the number of SP cells and metastasis. Increased miR-9 was detected in metastatic human primary SCCs and SCC metastases, and miR-9-transduced human SCC cells exhibited increased invasion. We identified α-catenin as a predominant miR-9 target. Increased miR-9 in human SCC metastases correlated with α-catenin loss but not E-cadherin loss. Our results demonstrate that stem cells with Kras(G12D) activation and Smad4 depletion can produce tumors that are multipotent and susceptible to EMT and metastasis. Additionally, tumor initiation and metastatic properties of CSCs can be uncoupled, with miR-9 regulating the expansion of metastatic CSCs.


Subject(s)
Carcinoma, Squamous Cell/secondary , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/genetics , Skin Neoplasms/pathology , Smad4 Protein/genetics , ras Proteins/genetics , Animals , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Cell Dedifferentiation , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mice, Transgenic , MicroRNAs/genetics , Mutation, Missense , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Proto-Oncogene Proteins p21(ras) , RNA Interference , Sequence Deletion , Side-Population Cells/metabolism , Side-Population Cells/pathology , Side-Population Cells/physiology , Skin Neoplasms/genetics , Tumor Cells, Cultured , alpha Catenin/genetics , alpha Catenin/metabolism
14.
J Invest Dermatol ; 133(11): 2609-2616, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23648546

ABSTRACT

Smad4 loss occurs frequently in human skin squamous cell carcinoma (SCC), but it is unknown whether this loss increases UV-induced carcinogenesis, a major etiological factor in skin cancer. In the present study, mice with keratinocyte-specific Smad4 deletion (K14.Smad4(-/-)) and wild-type (WT) littermates were chronically UV-irradiated. Compared with WT, K14.Smad4(-/-) mice exhibited increased DNA damage and increased susceptibility to UV-induced skin cancer. Among genes involved in repairing UV-induced DNA damage, Excision repair cross-complementation group 1 (Ercc1) messenger RNA was significantly reduced in UV-treated K14.Smad4(-/-) skin compared with WT skin. Further analysis revealed that Smad4 loss confers reduced Snail binding to the Ercc1 regulatory elements, resulting in reduced Ercc1 transcription. Consistently, transient transfection of Snai1 into Smad4(-/-) keratinocytes led to increased repair of UV-induced DNA lesions. Transfection of Ercc1 into Smad4(-/-) keratinocytes restored repair of UV-induced DNA damage. Further, immunostaining revealed that the presence of Smad4 protein is associated with the presence of Snail and Ercc1 proteins in human skin SCC and precancerous actinic keratoses. Collectively, Smad4 loss-associated Snail reduction compromises Ercc1-mediated DNA repair, contributing to increased UV-induced skin carcinogenesis. Thus, we identified a role for Snail in UV-induced DNA repair.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/physiopathology , DNA-Binding Proteins/genetics , Endonucleases/genetics , Keratinocytes/physiology , Skin Neoplasms/physiopathology , Smad4 Protein/genetics , Animals , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/genetics , Cells, Cultured , DNA Repair/physiology , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endonucleases/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Keratinocytes/cytology , Keratinocytes/radiation effects , Male , Mice , Mice, Mutant Strains , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/physiopathology , RNA, Messenger/metabolism , Skin Neoplasms/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Ultraviolet Rays/adverse effects
15.
Nat Med ; 19(4): 421-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23475202

ABSTRACT

We report that K5.Smad7 mice, which express a Smad7 transgene under the control of a keratin 5 promoter, were resistant to radiation-induced oral mucositis, a painful oral ulceration. In addition to nuclear factor κB (NF-κB) activation, which is known to contribute to oral mucositis, we found activated transforming growth factor ß (TGF-ß) signaling in cells from this condition. Smad7 dampened both pathways to attenuate inflammation, growth inhibition and apoptosis. Additionally, Smad7 promoted oral epithelial migration to close the wound. Further analyses revealed that TGF-ß signaling Smads and their co-repressor C-terminal binding protein 1 (CtBP1) transcriptionally repressed Rac1, and that Smad7 abrogated this repression. Knocking down Rac1 expression in mouse keratinocytes abrogated Smad7-induced migration. Topical application of Smad7 protein conjugated with a cell-permeable Tat tag to oral mucosa showed prophylactic and therapeutic effects on radiation-induced oral mucositis in mice. Thus, we have identified new molecular mechanisms involved in oral mucositis pathogenesis, and our data suggest an alternative therapeutic strategy to block multiple pathological processes in this condition.


Subject(s)
Radiation Injuries, Experimental/prevention & control , Smad7 Protein/physiology , Stomatitis/prevention & control , Alcohol Oxidoreductases/physiology , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Movement/physiology , DNA-Binding Proteins/physiology , Keratinocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/physiology , Neuropeptides/physiology , Signal Transduction/physiology , Smad7 Protein/genetics , Transforming Growth Factor beta/physiology , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein
16.
J Invest Dermatol ; 133(5): 1294-301, 2013 May.
Article in English | MEDLINE | ID: mdl-23303449

ABSTRACT

Carboxyl-terminal binding protein 1 (CtBP1) has been shown to suppress the transcription of several tumor suppressors in vitro. Paradoxically, a previous report showed that CtBP1 mRNA was downregulated in melanoma. Using immunostaining, we found that a large percentage of human melanomas were positive for CtBP1 protein. Furthermore, we demonstrated that CtBP1 expression in melanoma cells contributes to cell proliferation and genome instability, two aspects promoting melanoma initiation and progression. Breast cancer susceptibility gene 1 (Brca1), a core protein in DNA-damage repair, was repressed by CtBP1 in melanoma cells. Consistently, Brca1 loss in human malignant melanoma tissues was found to be inversely correlated with CtBP1 expression levels. In addition, the inhibitor of cyclin-dependent protein kinases (CDKs), p16INK4a, whose loss has been related to the pathogenesis of melanoma, was repressed by CtBP1 as well. Our findings suggest an important role of CtBP1 in the transcriptional control of p16INK4a and Brca1, with CtBP1 overexpression potentially contributing to increased proliferation and DNA damage in melanoma.


Subject(s)
Alcohol Oxidoreductases/metabolism , BRCA1 Protein/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Melanoma/metabolism , Skin Neoplasms/metabolism , Transcription, Genetic/physiology , Adult , Aged , Alcohol Oxidoreductases/genetics , BRCA1 Protein/genetics , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Disease Progression , Female , Humans , Male , Melanoma/pathology , Middle Aged , Skin Neoplasms/pathology
17.
J Immunol ; 190(2): 678-88, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23233723

ABSTRACT

Psoriasis is characterized by a specific microRNA expression profile, distinct from that of healthy skin. MiR-31 is one of the most highly overexpressed microRNAs in psoriasis skin; however, its biological role in the disease has not been studied. In this study, we show that miR-31 is markedly overexpressed in psoriasis keratinocytes. Specific inhibition of miR-31 suppressed NF-κB-driven promoter luciferase activity and the basal and TNF-α-induced production of IL-1ß, CXCL1/growth-related oncogene-α, CXCL5/epithelial-derived neutrophil-activating peptide 78, and CXCL8/IL-8 in human primary keratinocytes. Moreover, interference with endogenous miR-31 decreased the ability of keratinocytes to activate endothelial cells and attract leukocytes. By microarray expression profiling, we identified genes regulated by miR-31 in keratinocytes. Among these genes, we identified serine/threonine kinase 40 (STK40), a negative regulator of NF-κB signaling, as a direct target for miR-31. Silencing of STK40 rescued the suppressive effect of miR-31 inhibition on cytokine/chemokine expression, indicating that miR-31 regulates cytokine/chemokine expression via targeting STK40 in keratinocytes. Finally, we demonstrated that TGF-ß1, a cytokine highly expressed in psoriasis epidermis, upregulated miR-31 expression in keratinocytes in vitro and in vivo. Collectively, our findings suggest that overexpression of miR-31 contributes to skin inflammation in psoriasis lesions by regulating the production of inflammatory mediators and leukocyte chemotaxis to the skin. Our data indicate that inhibition of miR-31 may be a potential therapeutic option in psoriasis.


Subject(s)
Cytokines/biosynthesis , Gene Expression , Keratinocytes/metabolism , MicroRNAs/genetics , Protein Serine-Threonine Kinases/metabolism , Psoriasis/genetics , Psoriasis/immunology , Chemokines/biosynthesis , Chemotaxis, Leukocyte/immunology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , NF-kappa B , Protein Serine-Threonine Kinases/genetics , Psoriasis/enzymology , RNA Interference , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
18.
Int J Biol Sci ; 8(2): 228-35, 2012.
Article in English | MEDLINE | ID: mdl-22253566

ABSTRACT

TGFß1 was initially identified as a potent chemotactic cytokine to initiate inflammation, but the autoimmune phenotype seen in TGFß1 knockout mice reversed the dogma of TGFß1 being a pro-inflammatory cytokine to predominantly an immune suppressor. The discovery of the role of TGFß1 in Th17 cell activation once again revealed the pro-inflammatory effect of TGFß1. We developed K5.TGFß1 mice with latent human TGFß1 overexpression targeted to epidermal keratinocytes by keratin 5. These transgenic mice developed significant skin inflammation. Further studies revealed that inflammation severity correlated with switching TGFß1 transgene expression on and off, and genome wide expression profiling revealed striking similarities between K5.TGFß1 skin and human psoriasis, a Th1/Th17-associated inflammatory skin disease. Our recent study reveals that treatments alleviating inflammatory skin phenotypes in this mouse model reduced Th17 cells, and antibodies against IL-17 also lessen the inflammatory phenotype. Examination of inflammatory cytokines/chemokines affected by TGFß1 revealed predominantly Th1-, Th17-related cytokines in K5.TGFß1 skin. However, the finding that K5.TGFß1 mice also express Th2-associated inflammatory cytokines under certain pathological conditions raises the possibility that deregulated TGFß signaling is involved in more than one inflammatory disease. Furthermore, activation of both Th1/Th17 cells and regulatory T cells (Tregs) by TGFß1 reversely regulated by IL-6 highlights the dual role of TGFß1 in regulating inflammation, a dynamic, context and organ specific process. This review focuses on the role of TGFß1 in inflammatory skin diseases.


Subject(s)
Psoriasis/immunology , Transforming Growth Factor beta1/physiology , Animals , Gene Expression Profiling , Humans , Lymphocyte Activation , Mice , Mice, Transgenic , Models, Immunological , Psoriasis/genetics , Psoriasis/pathology , Th17 Cells/immunology
19.
Mol Carcinog ; 51(6): 500-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21681822

ABSTRACT

Carboxyl-terminal binding protein 1 (CtBP1) is a transcriptional co-repressor with oncogenic potential. Immunohistochemistry staining using human breast cancer tissue arrays revealed that 92% of invasive ductal breast cancer cases have CtBP1-positive staining compared to 4% CtBP1-positive in normal breast tissue. To explore the functional impact of CtBP1 in breast cancer, we examined CtBP1's transcriptional regulation of known tumor suppressors, breast cancer susceptibility gene 1 (Brca1), and E-cadherin. We found CtBP1 was recruited to the promoter regions of Brca1 and E-cadherin genes in breast cancer cells. Concomitantly, Brca1 loss was detected in 57% and E-cadherin loss was detected in 76% of human invasive ductal breast cancers, and correlated with CtBP1 nuclear staining in these lesions. Importantly, siRNA knock down of CtBP1 restored Brca1 and E-cadherin expression in breast cancer cell lines, implying CtBP1 down-regulates Brca1 and E-cadherin genes in human breast cancer. This study provides evidence that although genetic loss of Brca1 and E-cadherin are infrequent in breast cancer, they are down-regulated at the transcriptional level by CtBP1 expression. Thus, CtBP1 activation could be a potential biomarker for breast cancer development.


Subject(s)
Alcohol Oxidoreductases/metabolism , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Transcription, Genetic , Alcohol Oxidoreductases/genetics , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Promoter Regions, Genetic
20.
Cell Biosci ; 1: 41, 2011 Dec 28.
Article in English | MEDLINE | ID: mdl-22204491

ABSTRACT

Smad proteins are classified in different groups based on their functions in mediating transforming growth factor ß (TGFß) superfamily components. Smad1/5/8 mainly mediate bone morphogenetic proteins (BMP) pathway and Smad2/3 mainly mediate TGFß pathway. Smad4 functions as common Smad to mediate both pathways. Previous studies showed many members of TGFß superfamily play a role in carcinogenesis. The current review focuses on the role of TGFß signaling Smads in squamous cell carcinomas (SCCs). TGFß signaling inhibits early tumor development, but promotes tumor progression in the late stage. Although Smad2, Smad3 and Smad4 are all TGFß signaling Smads, they play different roles in SCCs. Genetically, Smad2 and Smad4 are frequently mutated or deleted in certain human cancers whereas Smad3 mutation or deletion is infrequent. Genetically engineered mouse models with these individual Smad deletions have provided important tools to identify their diversified roles in cancer. Using these models, we have shown that Smad4 functions as a potent tumor suppressor and its loss causes spontaneous SCCs development; Smad2 functions as a tumor suppressor and its loss promotes SCC formation initiated by other genetic insults but is insufficient to initiate tumor formation. In contrast, Smad3 primarily mediates TGFß-induced inflammation. The functions of each Smad also depends on the presence/absence of its Smad partner, thus need to be interpreted in a context-specific manner.

SELECTION OF CITATIONS
SEARCH DETAIL
...