Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Eur Acad Dermatol Venereol ; 37(2): 328-339, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36366861

ABSTRACT

PURPOSE: Cutaneous toxicities are common adverse effects following epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) therapy. Zinc deficiency causes diverse diseases, including skin toxicities. Therefore, this study aimed to investigate the role of zinc deficiency in patients with EGFR-TKI-induced skin toxicities. EXPERIMENTAL DESIGN: This retrospective study enrolled 269 patients with diverse skin disorders who visited our hospital between January 2016 and December 2017. The skin toxicity severities and plasma zinc levels of 101 EGFR-TKI-treated cancer patients were analysed and compared with those of 43 non-EGFR-TKI-treated cancer patients and 125 patients without cancer but presenting cutaneous manifestations. Additionally, the role of zinc in erlotinib-induced skin eruptions was established in a 14-day-murine model. Clinical features were further evaluated following systemic zinc supplementation in EGFR-TKI-treated cancer patients. RESULTS: EGFR-TKI-treated patients demonstrated severe cutaneous manifestations and a significant decrease in plasma zinc levels than those of the control groups. The serum zinc level and Common Terminology Criteria for Adverse Events (CTCAE) 5.0 grading of EGFR-TKI-induced skin toxicities showed a significant negative correlation (r = -0.29; p < 0.0001). Moreover, erlotinib treatment decreased the plasma zinc levels and induced periorificial dermatitis in rats confirming zinc deficiency following EGFR-TKI treatment. Zinc supplementation to the EGFR-TKI-treated cancer patients showed a significant decrease in the CTCEA grading (p < 0.0005 for mucositis and p < 0.0.0001 for all other cases) after 8 weeks. CONCLUSIONS: Skin impairment following EGFR-TKI therapy could be ameliorated through zinc supplementation. Thus, zinc supplementation should be considered for cancer patients undergoing EGFR-TKI therapy.


Subject(s)
Adenocarcinoma of Lung , Exanthema , Lung Neoplasms , Zinc , Animals , Mice , Rats , Adenocarcinoma of Lung/drug therapy , ErbB Receptors , Erlotinib Hydrochloride/adverse effects , Exanthema/chemically induced , Exanthema/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Zinc/metabolism
2.
Int J Mol Sci ; 23(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35563195

ABSTRACT

Berberine exerts therapeutic effects in inflammation-associated diseases. In a lipopolysaccharide (LPS)-induced endotoxemic acute lung injury (ALI) rat model, berberine alleviated lung injury through different anti-inflammatory mechanisms; however, treatment effects on CX3CL1 expression and shedding remain to be examined. As these processes play important roles in promoting the binding of leukocytes to the endothelium, the CX3CL1/CX3CR1 axis and its related pathways may serve as potential targets for the clinical treatment of ALI. The anti-inflammatory effects of berberine were investigated in LPS-stimulated rats, human umbilical cord vein endothelial cells (HUVECs), and THP-1 monocytic cells. Cx3cl1 expression in rat pulmonary tissues was examined using immunohistochemistry. CX3CL1, CX3CR1, RELA, STAT3, and ADAM10 levels were examined using Western blotting. CX3CL1 and ADAM10 mRNA levels were examined using quantitative real-time polymerase chain reaction. Soluble fractalkine levels in LPS-stimulated rats and HUVECs were examined using the enzyme-linked immunosorbent assay. Berberine significantly mitigated the LPS-induced upregulation of fractalkine and soluble fractalkine in rats and cultured HUVECs. Berberine mitigated the LPS-induced activation of the NF-κB and STAT3 signaling pathways. In THP-1 cells, berberine mitigated the LPS-induced upregulation of CX3CR1. Furthermore, the membrane expression of ADAM10 in LPS-stimulated HUVECs was suppressed by the berberine treatment. Berberine dose-dependently inhibited the LPS-induced activation of the CX3CL1/CX3CR1 axis and fractalkine shedding through ADAM10. These findings reveal a novel molecular mechanism underlying the inhibitory effect of berberine on monocyte adherence to the endothelium during inflammation.


Subject(s)
Acute Lung Injury , Berberine , ADAM10 Protein/genetics , Animals , Anti-Inflammatory Agents , Berberine/pharmacology , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Leukocytes/metabolism , Lipopolysaccharides/toxicity , Rats
3.
Int J Mol Sci ; 23(11)2022 Jun 03.
Article in English | MEDLINE | ID: mdl-35682967

ABSTRACT

Chemotherapy-induced thrombocytopenia (CIT) is a common complication when treating malignancies with cytotoxic agents wherein carboplatin is one of the most typical agents causing CIT. Janus kinase 2 (JAK2) is one of the critical enzymes to megakaryocyte proliferation and differentiation. However, the role of the JAK2 in CIT remains unclear. In this study, we used both carboplatin-induced CIT mice and MEG-01 cell line to examine the expression of JAK2 and signal transducer and activator of transcription 3 (STAT3) pathway. Under CIT, the expression of JAK2 was significantly reduced in vivo and in vitro. More surprisingly, the JAK2/STAT3 pathway remained inactivated even when thrombopoietin (TPO) was administered. On the other hand, carboplatin could cause prominent S phase cell cycle arrest and markedly increased apoptosis in MEG-01 cells. These results showed that the thrombopoiesis might be interfered through the downregulation of JAK2/STAT3 pathway by carboplatin in CIT, and the fact that exogenous TPO supplement cannot reactivate this pathway.


Subject(s)
Megakaryocytes , Thrombocytopenia , Animals , Apoptosis , Carboplatin/adverse effects , Cell Cycle Checkpoints , Down-Regulation , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Megakaryocytes/metabolism , Mice , S Phase , STAT3 Transcription Factor/metabolism , Signal Transduction , Thrombocytopenia/chemically induced , Thrombocytopenia/metabolism , Thrombopoietin/metabolism
4.
Int J Mol Sci ; 23(15)2022 Aug 07.
Article in English | MEDLINE | ID: mdl-35955918

ABSTRACT

Lidocaine injection is a common treatment for tendon injuries. However, the evidence suggests that lidocaine is toxic to tendon cells. This study investigated the effects of lidocaine on cultured tendon cells, focusing on the molecular mechanisms underlying cell proliferation and extracellular matrix (ECM) production. Tendon cells cultured from rat Achilles tendons were treated with 0.5, 1.0, or 1.5 mg/mL lidocaine for 24 h. Cell proliferation was evaluated by Cell Counting Kit 8 (CCK-8) assay and bromodeoxyuridine (BrdU) assay. Cell apoptosis was assessed by Annexin V and propidium iodide (PI) stain. Cell cycle progression and cell mitosis were assessed through flow cytometry and immunofluorescence staining, respectively. The expression of cyclin E, cyclin A, cyclin-dependent kinase 2 (CDK2), p21, p27, p53, matrix metalloproteinases-2 (MMP-2), matrix metalloproteinases-9 (MMP-9), type I collagen, and type III collagen were examined through Western blotting, and the enzymatic activity of MMP-9 was determined through gelatin zymography. Lidocaine reduced cell proliferation and reduced G1/S transition and cell mitosis. Lidocaine did not have a significant negative effect on cell apoptosis. Lidocaine significantly inhibited cyclin A and CDK2 expression but promoted p21, p27, and p53 expression. Furthermore, the expression of MMP-2 and MMP-9 increased, whereas that of type I and type III collagen decreased. Lidocaine also increased the enzymatic activity of MMP-9. Our findings support the premise that lidocaine inhibits tendon cell proliferation by changing the expression of cell-cycle-related proteins and reduces ECM production by altering levels of MMPs and collagens.


Subject(s)
Collagen Type III , Matrix Metalloproteinase 9 , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation , Collagen Type III/genetics , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Extracellular Matrix/metabolism , Lidocaine/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Rats , Tendons/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768826

ABSTRACT

Obstructive sleep apnea (OSA) is a disease with great cardiovascular risk. Interleukin-8 (IL-8), an important chemokine for monocyte chemotactic migration, was studied under intermittent hypoxia condition and in OSA patients. Monocytic THP-1 cells were used to investigate the effect of intermittent hypoxia on the regulation of IL-8 by an intermittent hypoxic culture system. The secreted protein and mRNA levels were studied by means of enzyme-linked immunosorbent assay and RT/real-time PCR. The chemotactic migration of monocytes toward a conditioned medium containing IL-8 was performed by means of the transwell filter migration assay. Peripheral venous blood was collected from 31 adult OSA patients and RNA was extracted from the monocytes for the analysis of IL-8 expression. The result revealed that intermittent hypoxia enhanced the monocytic THP-1 cells to actively express IL-8 at both the secreted protein and mRNA levels, which subsequently increased the migration ability of monocytes toward IL-8. The ERK, PI3K and PKC pathways were demonstrated to contribute to the activation of IL-8 expression by intermittent hypoxia. In addition, increased monocytic IL-8 expression was found in OSA patients, with disease severity dependence and diurnal changes. This study concluded the monocytic IL-8 gene expression can be activated by intermittent hypoxia and increased in OSA patients.


Subject(s)
Hypoxia/metabolism , Interleukin-8/biosynthesis , Sleep Apnea, Obstructive/metabolism , Adult , Female , Gene Expression , Humans , Hypoxia/genetics , Hypoxia/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Interleukin-8/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/genetics , Sleep Apnea, Obstructive/genetics , Sleep Apnea, Obstructive/immunology , THP-1 Cells
6.
Molecules ; 19(11): 19066-77, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25415472

ABSTRACT

BPC 157, a pentadecapeptide derived from human gastric juice, has been demonstrated to promote the healing of different tissues, including skin, muscle, bone, ligament and tendon in many animal studies. However, the underlying mechanism has not been fully clarified. The present study aimed to explore the effect of BPC 157 on tendon fibroblasts isolated from Achilles tendon of male Sprague-Dawley rat. From the result of cDNA microarray analysis, growth hormone receptor was revealed as one of the most abundantly up-regulated genes in tendon fibroblasts by BPC 157. BPC 157 dose- and time-dependently increased the expression of growth hormone receptor in tendon fibroblasts at both the mRNA and protein levels as measured by RT/real-time PCR and Western blot, respectively. The addition of growth hormone to BPC 157-treated tendon fibroblasts dose- and time-dependently increased the cell proliferation as determined by MTT assay and PCNA expression by RT/real-time PCR. Janus kinase 2, the downstream signal pathway of growth hormone receptor, was activated time-dependently by stimulating the BPC 157-treated tendon fibroblasts with growth hormone. In conclusion, the BPC 157-induced increase of growth hormone receptor in tendon fibroblasts may potentiate the proliferation-promoting effect of growth hormone and contribute to the healing of tendon.


Subject(s)
Achilles Tendon/drug effects , Achilles Tendon/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Peptide Fragments/pharmacology , Proteins/pharmacology , Receptors, Somatotropin/metabolism , Animals , Cell Proliferation/drug effects , Growth Hormone/metabolism , Janus Kinase 2/metabolism , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Wound Healing/drug effects
7.
J Orthop Res ; 42(1): 32-42, 2024 01.
Article in English | MEDLINE | ID: mdl-37442643

ABSTRACT

Muscle injuries are common among athletes and often treated with platelet-rich plasma (PRP). However, whether the leukocyte concentration affects the efficacy of PRP in treating muscle injuries remains unclear. This study investigated the effects of leukocyte-poor platelet-rich plasma (LP-PRP) and leukocyte-rich platelet-rich plasma (LR-PRP) on myoblast proliferation and the molecular mechanisms underlying these effects. Myoblasts were treated with 0.5% LP-PRP, 0.5% LR-PRP, 1% LP-PRP, or 1% LR-PRP for 24 h. The gene expression of the LP-PRP- and LR-PRP-treated myoblasts was determined using RNA sequencing analysis. Cell proliferation was evaluated using an bromodeoxyuridine (BrdU) assay, and cell cycle progression was assessed through flow cytometry. The expression of cyclin A, cyclin-dependent kinase 1 (cdk1), and cdk2 was examined using Western blotting. The expression of myoblast determination protein 1 (MyoD1) was examined through Western blotting and immunofluorescence staining. The LP-PRP and LR-PRP both promoted the proliferation of myoblasts and increased differential gene expression of myoblasts. Moreover, the LP-PRP and LR-PRP substantially upregulated the expression of cyclin A, cdk1, and cdk2. MyoD1 expression was induced in the LP-PRP and LR-PRP-treated myoblasts. Our results corroborate the finding that LP-PRP and LR-PRP have similar positive effects on myoblast proliferation and MyoD1 expression.


Subject(s)
Cyclin A , Myoblasts , Platelet-Rich Plasma , Humans , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin A/metabolism , Leukocytes/physiology , Myoblasts/physiology , Platelet-Rich Plasma/metabolism , Up-Regulation
8.
Food Chem Toxicol ; 175: 113740, 2023 May.
Article in English | MEDLINE | ID: mdl-36958389

ABSTRACT

Epidemiological studies have reported an association between chronic cadmium (Cd) exposure and increased cardiovascular risk; however, their causal relationship remains unclear. The aim of this study is to explore the effects of Cd exposure on the cardiac and arterial systems in mice. According to the concentration of cadmium chloride in drinking water, male mice were randomly divided into control and low-dose and high-dose Cd exposure groups. The intervention duration was 12 weeks. In cardiac tissues, Cd exposure led to focal necrosis, myofibril disarray, perivascular and interstitial fibrosis, and disorganized sarcomere structures. Cd also induced the apoptosis of cardiomyocytes and increased the expression levels of matrix metalloproteinase (MMP)-2 and MMP-14 in cardiac tissues. In the arterial tissues, Cd exposure damaged the intimal and medial layers of the aorta. Cd further reduced the viability of aortic smooth muscle cells in vitro. This study provides evidence for the Cd-induced damage of the cardiovascular system, which may contribute to various cardiovascular diseases.


Subject(s)
Cadmium , Heart , Mice , Male , Animals , Cadmium/toxicity , Cadmium/metabolism , Cadmium Chloride/metabolism , Lung/metabolism , Aorta
9.
Life Sci ; 310: 121072, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36228773

ABSTRACT

AIMS: Radiation-induced liver disease (RILD) is the major complication for cancer patients after radiation therapy. We investigated the protective effects of BPC 157 peptide in reducing RILD. MATERIALS AND METHODS: Mice were irradiated with a single dose of 12 Gy to induce acute liver injury with or without oral BPC 157. Plasma levels of AST and ALT were determined. In vitro rat liver clone 9 cells and in vivo liver tissues were harvested for MTT assay, TUNEL assay, lipid staining, polypoid cell counts, Western blotting of caspase-3, PCNA, KLF-4 and HIF-2α, and immunocytochemistry for PCNA, KLF-4 and HIF-2α. SiRNAs were used to knockdown KLF-4. KEY FINDINGS: BPC 157 was firstly demonstrated to reduce RILD by decreasing plasma levels of AST and ALT, and inhibiting hydropic degeneration of liver. BPC 157 significantly decreased radiation-induced cell apoptosis, increased PCNA expression, promoted the expression of KLF4, decreased the radiation-induced hepatic lipid accumulation and HIF-2α expression both in mice liver and in clone 9 liver cells. The knockdown of KLF4 abolished the protective effect of BPC 157 on radiation-induced apoptosis and lipid accumulation in clone 9 liver cells, indicating that the protective effect of BPC 157 was mediated by KLF4 in liver cells. SIGNIFICANCE: The present study provided a good model for molecular mechanism underlying the acute RILD. BPC 157, as a stable pentadecapeptide that can be chemically synthesized and purified easily for research, together with its in vivo markedly protective effect made it worth of being investigated for future clinical application for RILD.


Subject(s)
Anti-Ulcer Agents , Chemical and Drug Induced Liver Injury, Chronic , Rats , Animals , Mice , Kruppel-Like Factor 4 , Up-Regulation , Proliferating Cell Nuclear Antigen , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Peptide Fragments/therapeutic use , Basic Helix-Loop-Helix Transcription Factors , Lipids , Anti-Ulcer Agents/pharmacology
10.
EBioMedicine ; 73: 103654, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34740104

ABSTRACT

BACKGROUND: Age-related changes affecting the ocular surface cause vision loss in the elderly. Cisd2 deficiency drives premature aging in mice as well as resulting in various ocular surface abnormalities. Here we investigate the role of CISD2 in corneal health and disease. METHODS: We studied the molecular mechanism underlying the ocular phenotypes brought about by Cisd2 deficiency using both Cisd2 knockout (KO) mice and a human corneal epithelial cell (HCEC) cell line carrying a CRISPR-mediated CISD2KO background. We also develop a potential therapeutic strategy that targets the Ca2+ signaling pathway, which has been found to be dysregulated in the corneal epithelium of subjects with ocular surface disease in order to extend the mechanistic findings into a translational application. FINDINGS: Firstly, in patients with corneal epithelial disease, CISD2 is down-regulated in their corneal epithelial cells. Secondly, using mouse cornea, Cisd2 deficiency causes a cycle of chronic injury and persistent repair resulting in exhaustion of the limbal progenitor cells. Thirdly, in human corneal epithelial cells, CISD2 deficiency disrupts intracellular Ca2+ homeostasis, impairing mitochondrial function, thereby retarding corneal repair. Fourthly, cyclosporine A and EDTA facilitate corneal epithelial wound healing in Cisd2 knockout mice. Finally, cyclosporine A treatment restores corneal epithelial erosion in patients with dry eye disease, which affects the ocular surface. INTERPRETATION: These findings reveal that Cisd2 plays an essential role in the cornea and that Ca2+ signaling pathways are potential targets for developing therapeutics of corneal epithelial diseases. FUNDING: This study was supported by the Ministry of Science and Technology (MOST) and Chang Gung Medical Research Foundation, Taiwan.


Subject(s)
Epithelium, Corneal/physiology , Membrane Proteins/genetics , Regeneration , Animals , Biomarkers , Calcium/metabolism , Cell Line , Computational Biology/methods , Cyclosporine/pharmacology , Epithelial Cells/metabolism , Epithelium, Corneal/cytology , Female , Gene Expression Profiling , Homeostasis , Humans , Leukocytes/immunology , Leukocytes/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Imaging , Oxygen/metabolism , Regeneration/drug effects , Regeneration/genetics , Wound Healing/drug effects
11.
Materials (Basel) ; 14(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805683

ABSTRACT

The objectives of this work aim to investigate the interaction and cytotoxicity between nanometric graphene oxide (GO) and nasopharyngeal carcinoma cells (NPC-BM1), and possible application in photon therapy. GO nanosheets were obtained in the size range of 100-200 nm, with a negative surface charge. This nanometric GO exhibited a limited (<10%) cytotoxicity effect and no significant dimensional change on NPC-BM1 cells in the tested GO concentration range (0.1-10 µg·mL-1). However, the secondary protein structure was modified in the GO-treated NPC-BM1 cells, as determined through synchrotron radiation-based Fourier transform infrared microspectroscopy (SR-FTIRM) mapping. To further study the cellular response of GO-treated NPC-BM1 cancer cells at low GO concentration (0.1 µg·mL-1), photon radiation was applied with increasing doses, ranging from 2 to 8 Gy. The low radiation energy (<5 Gy) did not cause significant cell mortality (5-7%). Increasing the radiation energy to 6-8 Gy accelerated cell apoptosis rate, especially in the GO-treated NPC-BM1 cells (27%). This necrosis may be due to GO-induced conformational changes in protein and DNA/RNA, resulting in cell vulnerability under photon radiation. The findings of the present work demonstrate the potential biological applicability of nanometric GO in different areas, such as targeted drug delivery, cellular imaging, and radiotherapy, etc.

12.
Molecules ; 15(9): 6423-35, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20877233

ABSTRACT

The use of indigo naturalis to treat psoriasis has proved effective in our previous clinical studies. The present study was designed to examine the anti-inflammatory effect of indigo naturalis in primary cultured human umbilical vein endothelial cells (HUVECs). Pretreatment of cells with indigo naturalis extract attenuated TNF-α-induced increase in Jurkat T cell adhesion to HUVECs as well as decreased the protein and messenger (m)RNA expression levels of vascular cell adhesion molecule-1 (VCAM-1) on HUVECs. Indigo naturalis extract also inhibited the protein expression of activator protein-1 (AP-1)/c-Jun, a critical transcription factor for the activation of VCAM-1 gene expression. Since the reduction of lymphocyte adhesion to vascular cells by indigo naturalis extract could subsequently reduce the inflammatory reactions caused by lymphocyte infiltration in the epidermal layer and help to improve psoriasis, this study provides a potential mechanism for the anti-inflammatory therapeutic effect of indigo naturalis extract in psoriasis.


Subject(s)
Endothelial Cells/drug effects , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Tumor Necrosis Factor-alpha/physiology , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/analysis , Cell Adhesion/drug effects , Humans , Inflammation/drug therapy , Jurkat Cells/drug effects , Jurkat Cells/physiology , Psoriasis/drug therapy , RNA, Messenger/analysis , RNA, Messenger/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/genetics
13.
Nanomaterials (Basel) ; 10(2)2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32093180

ABSTRACT

In this study, the physicochemical and surface properties of the GO-Ag composite promote a synergistic antibacterial effect towards both Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. Aureus) bacteria. GO-Ag NPs have a better bactericidal effect on E. coli (73%) and S. Aureus (98.5%) than pristine samples (pure Ag or GO). Transmission electron microscopy (TEM) confirms that the GO layers folded entire bacteria by attaching to the membrane through functional groups, while the Ag NPs penetrated the inner cell, thus damaging the cell membrane and leading to cell death. Cyclic voltammetry (CV) tests showed significant redox activity in GO-Ag NPs, enabling good catalytic performance towards H2O2 reduction. Strong reactive oxygen species (ROS) in GO-Ag NPs suggests that ROS might be associated with bactericidal activity. Therefore, the synergy between the physicochemical effect and ROS production of this material is proposed as the mechanism of its antibacterial activity.

14.
Oncogene ; 39(24): 4695-4710, 2020 06.
Article in English | MEDLINE | ID: mdl-32404985

ABSTRACT

The overexpression of SOX4 in various kinds of cancer cells was associated with poor prognosis for patients. The role of SOX4 in angiogenesis and tumor microenvironment modulation was recently documented in breast cancer but remains unclear in hepatocellular carcinoma (HCC). In our study, the clinical relevance of SOX4 overexpression in HCC and its role in the tumor microenvironment were investigated. The overexpression of SOX4 (SOX4high) in tumor lesions was associated with higher microvessel density (P = 0.012), tumor thrombosis formation (P = 0.012), distant metastasis (P < 0.001), and an independent prognostic factor for disease-free survival in HCC patients (P = 0.048). Endogenous SOX4 knockout in Hep3B cells by the CRISPR/cas9 system reduced the expression of CXCL12, which, in turn, attenuated chemotaxis in human umbilical vein endothelial cells, tube formation in vitro, reduced tumor growth, reticular fiber production, and angiogenesis in vivo in a xenograft mouse model. Treatment with an antagonist targeting CXCR4 (AMD3100), a receptor of CXCL12, inhibited chemotaxis and tube formation in endothelial cells in vitro. The CXCL12 promoter was activated by ectopic expression of a Flag-tagged SOX4 plasmid, endogenous SOX4 knockdown abolished promoter activity of CXCL12 as shown by luciferase assays, and an association with the CXCL12 promoter was identified via chromatin immunoprecipitation in HCC cells. In conclusion, SOX4 modulates the CXCL12 promoter in HCC cells. The secretory CXCL12, in turn, modulates CXCR4 in endothelial cells, reticular fibers to regulate the tumor microenvironment and modulate neovascularization, which might contribute to the distant metastasis of tumors.


Subject(s)
Carcinoma, Hepatocellular , Cell Movement , Chemokine CXCL12/metabolism , Human Umbilical Vein Endothelial Cells , Liver Neoplasms, Experimental , Neoplasm Proteins , Neovascularization, Pathologic , SOXC Transcription Factors/metabolism , Thrombosis , Animals , CRISPR-Cas Systems , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemokine CXCL12/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , SOXC Transcription Factors/genetics , Thrombosis/genetics , Thrombosis/metabolism , Thrombosis/pathology , Tumor Microenvironment/genetics
15.
Int J Dermatol ; 59(3): 326-332, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31763692

ABSTRACT

BACKGROUND: Paronychia is a common adverse event caused by epidermal growth factor receptor (EGFR) inhibitors. However, high rates of post-treatment discomfort, infection, recurrence, and increased time to return to work have been noted after nail plate avulsion for EGFR inhibitor-induced paronychia. Furthermore, poor wound healing and malnutrition were common conditions found in cancer patients. The aim of this study is to find an effective, pain-relieving, and noninvasive treatment for patients with severe paronychia induced by EGFR inhibitors. METHODS: Data from a series of 35 non-small cell lung cancer cases suffering from EGFR inhibitor-induced paronychia with pyogenic granuloma-like lesions of digits treated with betaxolol 0.25% ophthalmic solution once daily were collected and analyzed. RESULTS: Of the 35 patients suffering from grade 2 or 3 paronychia with pyogenic granuloma-like lesions induced by EGFR inhibitors, 34 (97.1%) demonstrated complete resolution and only one (2.9%) had partial resolution after 12 weeks of topical betaxolol treatment. The grading of paronychia according to the Common Terminology Criteria for Adverse Events decreased from an average of 2.29 to 0.63 after 4 weeks of treatment (P = 5.55 × 10-16 ). All the patients had significant improvement (50% pain reduction), as their pain visual analogue scale scores decreased from an average of 7.06 to 2.26 after one week of treatment (P = 6.11 × 10-25 ). CONCLUSION: Betaxolol 0.25% ophthalmic solution is an effective, safe, and pain-relieving treatment for patients suffering from EGFR inhibitor-induced paronychia with pyogenic granuloma-like lesions and deep fissures.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/administration & dosage , Betaxolol/administration & dosage , Dermatologic Agents/administration & dosage , Granuloma, Pyogenic/drug therapy , Paronychia/drug therapy , Protein Kinase Inhibitors/adverse effects , Administration, Topical , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Female , Granuloma, Pyogenic/chemically induced , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Paronychia/chemically induced , Protein Kinase Inhibitors/therapeutic use , Wound Healing/drug effects
16.
Free Radic Biol Med ; 120: 266-276, 2018 05 20.
Article in English | MEDLINE | ID: mdl-29609021

ABSTRACT

OBJECTIVE: Smoking and high-sensitivity C-reactive protein (hs-CRP) are risk factors for coronary artery spasm (CAS), which is characterized by the increased interleukin-6 (IL-6) level and monocyte counts; however, limited data are available regarding the role of cigarette-embedded nicotine in the modulation of monocytic inflammatory activity in CAS. APPROACH: We investigated and elucidated the putative roles and associations of nicotine, monocytic IL-6, α7 nicotinic acetylcholine receptor (α7-nAChR), and CRP in CAS development. RESULTS: We demonstrated that a significantly increased α7-nAChR (p = 0.001) and IL-6 (p = 0.0036) messenger RNA (mRNA) expression in the serum of patients with CAS. Serum hs-CRP levels exhibited a strong positive correlation with the monocytic mRNA expression of α7-nAChR (r = 0.71, p < 0.001) and IL-6 (r = 0.49, p = 0.006). The α7-nAChR and IL-6 expression levels of the CAS group were also positively correlated (r = 0.63, p < 0.001). Compared with the untreated controls, THP-1 cells and patient-derived monocytes treated with different concentrations of CRP displayed significantly increased expression levels of α7-nAChR mRNA and protein (p = 0.0054), in a dose-dependent manner. We also demonstrated that compared with the IL-6 expression elicited by CRP alone (p = 0.0489), the CRP-induced rise in monocytic IL-6 mRNA and protein expression in the presence of nicotine (p = 0.0002), is mediated by α7-nAChR activation and the deregulation of the human p38 mitogen-activated protein kinases (MAPK) signaling pathway. CONCLUSIONS: Our data demonstrate that the elevated monocytic IL-6 and α7-nAChR mRNA and protein expression levels are associated with the interaction between nicotine and CRP positively modulates CAS development. Our study suggests the potential role of α7-nAChR mRNA and/or protein expression as a diagnostic biomarker for CAS.


Subject(s)
Coronary Vasospasm/metabolism , MAP Kinase Signaling System/physiology , Monocytes/metabolism , Oxidative Stress/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Aged , C-Reactive Protein/metabolism , Cohort Studies , Coronary Vasospasm/physiopathology , Female , Humans , Inflammation/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Oxidative Stress/drug effects , Prospective Studies , Smoking/adverse effects
17.
Invest Ophthalmol Vis Sci ; 48(12): 5699-707, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18055822

ABSTRACT

PURPOSE: To investigate whether overexpression of the cytoprotective gene heme oxygenase-1 (HO-1) in photoreceptors by gene delivery attenuates cellular injury caused by intense light damage and to document the possible mechanisms of protection. METHODS: Recombinant adeno-associated virus type 5 (rAAV5) expressing the mouse HO-1 gene (mHO-1) was delivered to cyclic-light reared Sprague-Dawley (SD) rats by subretinal injection. Three weeks after transfer of HO-1 gene, animals were subjected to 2-hour intense light exposure then were returned to darkness. Expression of HO-1, p53, p38, and cellular FLICE inhibitory protein (c-FLIP) at different times after intense light damage was evaluated by Western blot analysis. HO-1 transgene expression, along with expression of c-fos and bcl-2, was analyzed by immunohistochemistry. In addition, the protective effects of HO-1 were evaluated by determining the morphology of the retina. Finally, apoptosis in photoreceptors was measured using TdT-dUTP terminal nick-end labeling (TUNEL) 24 hours after photic injury. RESULTS: Exogenous administration of HO-1 by gene transfer led to HO-1 transgene expression in photoreceptors. Protection of retina by HO-1 overexpression is evident from the partially preserved retina structure and attenuated apoptosis in photoreceptors after photic injury. Concurrently, overexpression of HO-1 was associated with a decrease in the expression of c-fos and p53, an increase in the activation of p38 and bcl-2, and preserved the expression of c-FLIP. CONCLUSIONS: Overexpression of HO-1 in photoreceptors protected themselves from subsequent cellular damage caused by intense light exposure. The anti-apoptotic mechanisms of HO-1 may be related to the induction of p38, bcl-2, and c-FLIP and to the suppression of c-fos and p53.


Subject(s)
Dependovirus/genetics , Gene Expression Regulation, Enzymologic , Heme Oxygenase (Decyclizing)/genetics , Light/adverse effects , Photoreceptor Cells, Vertebrate/radiation effects , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/prevention & control , Animals , Blotting, Western , Caspase 8/metabolism , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Heme Oxygenase (Decyclizing)/metabolism , In Situ Nick-End Labeling , Male , Photoreceptor Cells, Vertebrate/ultrastructure , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Radiation Injuries, Experimental/etiology , Rats , Rats, Sprague-Dawley , Retinal Degeneration/etiology , Retinal Degeneration/genetics , Retinal Degeneration/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Cornea ; 25(1): 78-84, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16331046

ABSTRACT

PURPOSE: To investigate the effect of corneal epithelium on the viability of corneal stromal keratocytes in Optisol-GS. METHODS: After sterilization, corneoscleral buttons were excised and stored in Optisol-GS for various time periods. Group 1 corneas (n = 40) underwent mechanical corneal epithelial debridement before storage while group 2 corneas (n = 40) were stored with intact epithelium. Changes in corneal thickness, keratocyte density, and keratocyte apoptosis were investigated immediately, at 4 hours, and on days 1, 2, 3, 5, 7, and 14 in the preservation medium. The differences between group 1 and 2 corneas were analyzed. RESULTS: Corneal thickness increased significantly in the second week of preservation in both groups, though more substantially in group 1. Significant corneal epithelial apoptosis was noticed in the first week in group 2 corneas. Corneal stromal keratocyte density decreased with prolonged preservation time. DNA laddering was detected by ligation-mediated polymerase chain reaction throughout the experiment periods in both groups, but the increase of keratocyte apoptosis was more significant after 5 days of preservation, especially in group 1. CONCLUSIONS: Stromal keratocytes underwent apoptosis in Optisol-GS. The absence of corneal epithelium during preservation further increased the stromal keratocyte apoptosis.


Subject(s)
Apoptosis , Chondroitin Sulfates/pharmacology , Corneal Stroma/pathology , Dextrans/pharmacology , Epithelial Cells/physiology , Gentamicins/pharmacology , Organ Preservation Solutions/pharmacology , Organ Preservation , Animals , Cell Communication , Cell Count , Cell Survival , Complex Mixtures/pharmacology , Corneal Stroma/drug effects , Cryopreservation , DNA/analysis , Debridement , Epithelium, Corneal/cytology , Fibroblasts/pathology , In Situ Nick-End Labeling , Polymerase Chain Reaction , Swine
19.
Invest Ophthalmol Vis Sci ; 46(3): 808-15, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15728535

ABSTRACT

PURPOSE: To investigate the expression and pivotal role of matrix metalloproteinase (MMP)-9 in the ex vivo expansion of human limbal explants with or without amniotic membrane (AM). METHODS: Corneoscleral buttons were cultured on intact, denuded AM or plastic dishes for 3 weeks. To determine the role of MMP-9 in cell migration, either the MMP inhibitor GM6001 or an MMP-9 antibody was used. Expression of MMP-9 was determined by gelatin zymography, reverse transcription-polymerase chain reaction, and immunohistochemical staining. RESULTS: The expression of MMP-9 in all culture conditions increased in a time-dependent manner. However, the active form of MMP-9 emerged only in cultures on both intact and denuded AM from the second week. The averaged corrected ratio of MMP-9 expression in cultures on intact AM versus those on denuded AM or plastic dishes was 2.76 +/- 0.69- or 4.25 +/- 0.30-fold, respectively, when total RNA was used as an internal control. MMP-9 transcripts were upregulated in cultures on intact AM compared with the other two culture conditions. Immunohistochemical staining demonstrated that the MMP-9 protein was located on the limbal epithelial cells. Upregulation of MMP-9 associated with cell migration was significantly attenuated by both GM6001 and MMP-9 antibody, consistent with the inhibition of MMP-9 activity, as determined by gelatin zymography. In contrast, the sizes of limbal outgrowth were not different between the control and MMP-9 antibody-treated plastic dishes. CONCLUSIONS: These results demonstrated that MMP-9 not only was upregulated, it was also involved in the outgrowth of limbal epithelial cells. These results suggest that cell-cell matrix interaction is involved in the expansion of limbal epithelial cells on intact AM, and MMP-9 may be a key element.


Subject(s)
Amnion/cytology , Epithelial Cells/cytology , Epithelium, Corneal/cytology , Limbus Corneae/cytology , Matrix Metalloproteinase 9/physiology , Adolescent , Adult , Aged , Cell Movement , Cells, Cultured , Coculture Techniques , Dipeptides/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelium, Corneal/drug effects , Epithelium, Corneal/enzymology , Humans , Immunoenzyme Techniques , Limbus Corneae/enzymology , Matrix Metalloproteinase 2/metabolism , Middle Aged , Protease Inhibitors/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
20.
PLoS One ; 10(12): e0144571, 2015.
Article in English | MEDLINE | ID: mdl-26673160

ABSTRACT

Transplantation of ex vivo cultured limbal epithelial cells is proven effective in restoring limbal stem cell deficiency. The present study aimed to investigate the promoting effect of Y-27632 on limbal epithelial cell proliferation. Limbal explants isolated from human donor eyes were expanded three weeks on culture dishes and outgrowth of epithelial cells was subsequently subcultured for in vitro experiments. In the presence of Y-27632, the ex vivo limbal outgrowth was accelerated, particularly the cells with epithelial cell-like morphology. Y-27632 dose-dependently promoted the proliferation of in vitro cultured human limbal epithelial cells as examined by phase contrast microscopy and luminescent cell-viability assay 30 hours after the treatment. The colony forming efficacy determined 7 days after the treatment was enhanced by Y-27632 also in a dose-dependent manner. The number of p63- or Ki67-positive cells was dose-dependently increased in Y-27632-treated cultures as detected by immunofluorescent staining and western blotanalysis. Cell cycle analysis by flow cytometric method revealed an increase in S-phase proliferating cells. The epithelial woundclosure rate was shown to be faster in experimental group received topical treatment withY-27632 than the sham control using a rat corneal wounding model. These resultsdemonstrate that Y-27632 can promote both the ex vivo and in vitro proliferation oflimbal epithelial cell proliferation. The in vivo enhanced epithelial wound healingfurther implies that the Y-27632 may act as a new strategy for treating limbal stem cell deficiency.


Subject(s)
Amides/pharmacology , Epithelial Cells/pathology , Limbus Corneae/pathology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Wound Healing/drug effects , rho-Associated Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Keratin-12/metabolism , Ki-67 Antigen/metabolism , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL