Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928170

ABSTRACT

Reactive pustular eruptions (RPEs) can manifest in a variety of conditions, including pustular psoriasis (PP) and adult-onset immunodeficiency syndrome due to anti-interferon-γ autoantibody (AOID). These RPEs can be attributed to different causes, one of which is genetic factors. However, the genetic basis for pustular skin diseases remains poorly understood. In our study, we conducted whole-exome sequencing on a cohort of 17 AOID patients with pustular reactions (AOID-PR) and 24 PP patients. We found that 76% and 58% of the AOID-PR and PP patients, respectively, carried rare genetic variations within the filaggrin (FLG) gene family. A total of 12 out of 21 SNPs on FLG had previously received clinical classifications, with only p.Ser2706Ter classified as pathogenic. In contrast, none of the FLG3 SNPs identified in this study had prior clinical classifications. Overall, these variations had not been previously documented in cases of pustular disorders, and two of them were entirely novel discoveries. Immunohistochemical analysis of skin biopsies revealed that FLG variants like p.Ser860Trp, p.Gly3903Ter, p.Gly2440Glu, and p.Glu2133Asp caused reductions in FLG levels similar to the pathogenic FLG p.Ser2706Ter. These results highlight rare FLG variants as potential novel genetic risk factors contributing to pustule formation in both AOID and PP.


Subject(s)
Asian People , Filaggrin Proteins , Intermediate Filament Proteins , Polymorphism, Single Nucleotide , Humans , Intermediate Filament Proteins/genetics , Female , Male , Asian People/genetics , Adult , Middle Aged , Exome Sequencing , Genetic Predisposition to Disease , Psoriasis/genetics , Psoriasis/pathology , Aged , Interferon-gamma/genetics , Interferon-gamma/metabolism , Autoantibodies/immunology , Skin/pathology , Skin/metabolism
2.
Int J Mol Sci ; 25(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38279264

ABSTRACT

Hepatocellular carcinoma (HCC) presents a significant global health challenge due to limited early detection methods, primarily relying on conventional approaches like imaging and alpha-fetoprotein (AFP). Although non-coding RNAs (ncRNAs) show promise as potential biomarkers in HCC, their true utility remains uncertain. We conducted a comprehensive review of 76 articles, analyzing 88 circulating lncRNAs in 6426 HCC patients. However, the lack of a standardized workflow protocol has hampered holistic comparisons across the literature. Consequently, we herein confined our meta-analysis to only a subset of these lncRNAs. The combined analysis of serum highly upregulated in liver cancer (HULC) gene expression with homeobox transcript antisense intergenic RNA (HOTAIR) and urothelial carcinoma-associated 1 (UCA1) demonstrated markedly enhanced sensitivity and specificity in diagnostic capability compared to traditional biomarkers or other ncRNAs. These findings could have substantial implications for the early diagnosis and tailored treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Carcinoma, Transitional Cell , Liver Neoplasms , RNA, Long Noncoding , Urinary Bladder Neoplasms , Humans , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/pathology , RNA, Long Noncoding/metabolism , Genes, Homeobox , RNA, Antisense , Carcinoma, Transitional Cell/genetics , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/genetics , RNA, Untranslated , Biomarkers , Gene Expression Profiling , Biomarkers, Tumor/genetics
3.
Trends Cancer ; 9(8): 679-690, 2023 08.
Article in English | MEDLINE | ID: mdl-37230895

ABSTRACT

Helicobacter pylori is a human microbial pathogen that colonizes the gastric epithelium and causes type B gastritis with varying degrees of active inflammatory infiltrates. The underlying chronic inflammation induced by H. pylori and other environmental factors may promote the development of neoplasms and adenocarcinoma of the stomach. Dysregulation of various cellular processes in the gastric epithelium and in different cells of the microenvironment is a hallmark of H. pylori infection. We address the conundrum of H. pylori-associated apoptosis and review distinct mechanisms induced in host cells that either promote or suppress apoptosis in gastric epithelial cells, often simultaneously. We highlight key processes in the microenvironment that contribute to apoptosis and gastric carcinogenesis.


Subject(s)
Gastritis , Helicobacter pylori , Stomach Neoplasms , Humans , Stomach Neoplasms/etiology , Gastric Mucosa/pathology , Apoptosis , Tumor Microenvironment
4.
Biochim Biophys Acta Mol Cell Res ; 1869(12): 119364, 2022 12.
Article in English | MEDLINE | ID: mdl-36162648

ABSTRACT

Crosstalk within the gastric epithelium, which is closely in contact with stromal fibroblasts in the gastric mucosa, has a pivotal impact in proliferation, differentiation and transformation of the gastric epithelium. The human pathogen Helicobacter pylori colonises the gastric epithelium and represents a risk factor for gastric pathophysiology. Infection of H. pylori induces the activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which is involved in the pro-inflammatory response but also in cell survival. In co-cultures with human gastric fibroblasts (HGF), we found that apoptotic cell death is reduced in the polarised human gastric cancer cell line NCI-N87 or in gastric mucosoids during H. pylori infection. Interestingly, suppression of apoptotic cell death in NCI-N87 cells involved an enhanced A20 expression regulated by NF-κB activity in response to H. pylori infection. Moreover, A20 acts as an important negative regulator of caspase-8 activity, which was suppressed in NCI-N87 cells during co-culture with gastric fibroblasts. Our results provide evidence for NF-κB-dependent regulation of apoptotic cell death in cellular crosstalk and highlight the protective role of gastric fibroblasts in gastric epithelial cell death during H. pylori infection.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Caspase 8/metabolism , Cell Survival , Coculture Techniques , Epithelial Cells/metabolism , Fibroblasts/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Humans , NF-kappa B/metabolism , Transcription Factors/metabolism
5.
Cell Mol Life Sci ; 79(8): 461, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35913642

ABSTRACT

The human pathogen Helicobacter pylori represents a risk factor for the development of gastric diseases including cancer. The H. pylori-induced transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is involved in the pro-inflammatory response and cell survival in the gastric mucosa, and represents a trailblazer of gastric pathophysiology. Termination of nuclear NF-κB heterodimer RelA/p50 activity is regulated by the ubiquitin-RING-ligase complex elongin-cullin-suppressor of cytokine signalling 1 (ECSSOCS1), which leads to K48-ubiquitinylation and degradation of RelA. We found that deubiquitinylase (DUB) ubiquitin specific protease 48 (USP48), which interacts with the COP9 signalosome (CSN) subunit CSN1, stabilises RelA by deubiquitinylation and thereby promotes the transcriptional activity of RelA to prolong de novo synthesis of DUB A20 in H. pylori infection. An important role of A20 is the suppression of caspase-8 activity and apoptotic cell death. USP48 thus enhances the activity of A20 to reduce apoptotic cell death in cells infected with H. pylori. Our results, therefore, define a synergistic mechanism by which USP48 and A20 regulate RelA and apoptotic cell death in H. pylori infection.


Subject(s)
Helicobacter Infections , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Specific Proteases , Cell Survival , Helicobacter pylori , Humans , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
6.
Front Immunol ; 13: 866925, 2022.
Article in English | MEDLINE | ID: mdl-35663960

ABSTRACT

Background: Trauma is the third leading cause of mortality worldwide. Upon admission, up to 50% of traumatized patients are acutely intoxicated with alcohol, which might lead to aberrant immune responses. An excessive and uncontrolled inflammatory response to injury is associated with damage to trauma-distant organs. We hypothesize that, along with inflammation-induced apoptosis, the activation of the Wnt/ß-catenin signaling pathway would cause breakdown of the lung barrier and the development of lung injury after trauma. It remains unclear whether ethanol intoxication (EI) prior to trauma and hemorrhagic shock will attenuate inflammation and organ injury. Methods: In this study, 14 male C57BL/6J mice were randomly assigned to two groups and exposed either to EtOH or to NaCl as a control by an oral gavage before receiving a femur fracture (Fx) and hemorrhagic shock, followed by resuscitation (THFx). Fourteen sham animals received either EtOH or NaCl and underwent surgical procedures without THFx induction. After 24 h, oil red O staining of fatty vacuoles in the liver was performed. Histological lung injury score (LIS) was assessed to analyze the trauma-induced RLI. Gene expression of Cxcl1, Il-1ß, Muc5ac, Tnf, and Tnfrsf10b as well as CXCL1, IL-1ß, and TNF protein levels in the lung tissue and bronchoalveolar lavage fluid were determined by RT-qPCR, ELISA, and immunohistological analyses. Infiltrating polymorphonuclear leukocytes (PMNLs) were examined via immunostaining. Apoptosis was detected by activated caspase-3 expression in the lung tissue. To confirm active Wnt signaling after trauma, gene expression of Wnt3a and its inhibitor sclerostin (Sost) was determined. Protein expression of A20 and RIPK4 as possible modulators of the Wnt signaling pathway was analyzed via immunofluorescence. Results: Significant fatty changes in the liver confirmed the acute EI. Histopathology and decreased Muc5ac expression revealed an increased lung barrier breakdown and concomitant lung injury after THFx versus sham. EI prior trauma decreased lung injury. THFx increased not only the gene expression of pro-inflammatory markers but also the pulmonary infiltration with PMNL and apoptosis versus sham, while EI prior to THFx reduced those changes significantly. EI increased the THFx-reduced gene expression of Sost and reduced the THFx-induced expression of Wnt3a. While A20, RIPK4, and membranous ß-catenin were significantly reduced after trauma, they were enhanced upon EI. Conclusion: These findings suggest that acute EI alleviates the uncontrolled inflammatory response and lung barrier breakdown after trauma by suppressing the Wnt/ß-catenin signaling pathway.


Subject(s)
Alcoholic Intoxication , Lung Injury , Shock, Hemorrhagic , Animals , Disease Models, Animal , Ethanol/toxicity , Humans , Inflammation/pathology , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/pathology , Sodium Chloride , Wnt Signaling Pathway
7.
ACS Biomater Sci Eng ; 7(12): 5474-5483, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34704732

ABSTRACT

Microfluidic devices for culturing cells have been successfully utilized for biomedical applications, including drug screening. Several cell lines could be cultivated in microengineered environments with promising results, but gastric cell lines have not yet been widely used or studied. Therefore, this study focuses on establishing a polarized gastric epithelial monolayer on-a-chip and describes a general-purpose methodology applicable for bonding any porous material to PDMS through an adhesive sublayer. The fully transparent microfluidic chip consists of two microfluidic channels separated by a collagen-coated porous membrane and lined by human polarized gastric epithelial (NCI-N87) cells. We present considerations on how to ensure continuous and stable flow through the channels. The continuous flow rate was achieved using a pressure-driven pump. Media flow at a constant rate (0.5 µL/min) rapidly led the gastric epithelial cells to develop into a polarized monolayer. The barrier integrity was assessed by the FITC-dextran test. The generation of a monolayer was faster than in the static Boyden chamber. Moreover, fluorescence microscopy was used to monitor the apoptotic cell death of gastric epithelial monolayers on-a-chip in response to camptothecin, a therapeutic gastric cancer drug.


Subject(s)
Lab-On-A-Chip Devices , Pharmaceutical Preparations , Apoptosis , Epithelial Cells , Humans , Microfluidics
8.
Biomedicines ; 9(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34440074

ABSTRACT

Gastric cancer is considered one of the most common causes of cancer-related death worldwide and, thus, a major health problem. A variety of environmental factors including physical and chemical noxae, as well as pathogen infections could contribute to the development of gastric cancer. The transcription factor nuclear factor kappa B (NF-κB) and its dysregulation has a major impact on gastric carcinogenesis due to the regulation of cytokines/chemokines, growth factors, anti-apoptotic factors, cell cycle regulators, and metalloproteinases. Changes in NF-κB signaling are directed by genetic alterations in the transcription factors themselves, but also in NF-κB signaling molecules. NF-κB actively participates in the crosstalk of the cells in the tumor micromilieu with divergent effects on the heterogeneous tumor cell and immune cell populations. Thus, the benefits/consequences of therapeutic targeting of NF-κB have to be carefully evaluated. In this review, we address recent knowledge about the mechanisms and consequences of NF-κB dysregulation in gastric cancer development and therapy.

9.
Trends Biotechnol ; 39(8): 745-748, 2021 08.
Article in English | MEDLINE | ID: mdl-33353764

ABSTRACT

2D monolayer gastric organoids (2DMGOs)-on-a-chip have consistent structures and can live for more than a year in culture. This state-of-the-art cell physiological system in a microfluidic device provides a way to investigate biomedically relevant, stimuli-dependent cellular responses in a variety of differentiated 2DMGOs.


Subject(s)
Lab-On-A-Chip Devices , Organoids , Physiology , Stomach , Biomedical Research/instrumentation , Biomedical Research/methods , Humans , Organoids/physiology , Physiology/instrumentation , Physiology/methods , Stomach/physiology
10.
Biol Pharm Bull ; 41(11): 1659-1666, 2018.
Article in English | MEDLINE | ID: mdl-30381665

ABSTRACT

Lymphangiogenesis, the formation of lymphatic vessels from preexisting ones, promotes cancer growth and metastasis. Finding natural compounds with anti-lymphangiogenic activity will be useful for preventive treatment of lymphatic metastasis. Shikonin, an ingredient of a traditional Japanese and Chinese medicinal herb Lithospermum erythrorhizon, has been widely used in several pharmaceutical and cosmetic preparations, as well as in food colorants. Shikonin has been reported to inhibit lymphangiogenesis in vitro, but the mechanism of inhibition has not been determined. The aim of this study is to investigate the mechanism of anti-lymphangiogenesis of shikonin in primary human lymphatic endothelial cells (HMVEC-dLy). Shikonin, at non-toxic concentrations, significantly inhibited cord formation ability of lymphatic endothelial cells in a dose- and time-dependent manner. Western blotting analysis showed that shikonin decreased nuclear factor-kappaB (NF-κB) activation, as indicated by phosphorylation and nuclear translocation of NF-κB p65, and also reduced both mRNA and protein levels of hypoxia-inducible factor-1 (HIF-1)α. Use of an NF-κB inhibitor (BAY 11-7085) and HIF-1α small interfering RNA (siRNA) transfection revealed that NF-κB activation was upstream of HIF-1α expression, which controls cord formation by HMVEC-dLy. In addition, the reduction of vascular endothelial growth factor C (VEGF-C) and vascular endothelial growth factor receptor-3 (VEGFR-3) mRNA levels were also found in HMVEC-dLy that treated with shikonin. In conclusion, shikonin inhibits lymphangiogenesis in vitro by interfering the NF-κB/HIF-1α pathway and involves in suppression of VEGF-C and VEGFR-3 mRNA expression.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lithospermum/chemistry , Lymphangiogenesis/drug effects , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Drugs, Chinese Herbal/therapeutic use , Endothelial Cells/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphatic Metastasis/prevention & control , Naphthoquinones/therapeutic use , Phytotherapy , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
11.
Vasc Med ; 23(3): 201-211, 2018 06.
Article in English | MEDLINE | ID: mdl-29629844

ABSTRACT

Lymphangiogenesis is the process of new vessel formation from pre-existing lymphatic vessels. The process mainly involves cell adhesion, migration, and tubule formation of lymphatic endothelial cells. Tumor-induced lymphangiogenesis is an important factor contributing to promotion of tumor growth and cancer metastasis via the lymphatic system. Finding the non-toxic agents that can prevent or inhibit lymphangiogenesis may lead to blocking of lymphatic metastasis. Recently, aspirin, a non-steroidal anti-inflammatory drug (NSAID), has been reported to inhibit in vivo lymphangiogenesis in tumor and incision wound models, but the mechanisms of actions of aspirin on anti-lymphangiogenesis have been less explored. In this study, we aim to explore the mechanism underlying the anti-lymphangiogenic effects of aspirin in primary human dermal lymphatic microvascular endothelial (HMVEC-dLy) cells in vitro. Pretreatment of aspirin at non-toxic dose 0.3 mM significantly suppressed in vitro cord formation, adhesion, and the migration abilities of the HMVEC-dLy cells. Western blotting analysis indicated that aspirin decreased expression of vascular cell adhesion molecule-1 (VCAM-1), at both protein and mRNA levels, and these correlated with the reduction of NF-κB p65 phosphorylation. By using NF-κB inhibitor (BAY-11-7085) and VCAM-1 siRNA, we showed that VCAM-1 expression is downstream of NF-κB activation, and this NF-κB/VCAM-1 signaling pathway controls cord formation, adhesion, and the migration abilities of the HMVEC-dLy cells. In summary, we demonstrate the potential of aspirin as an anti-lymphangiogenic agent, and elucidate its mechanism of action.


Subject(s)
Aspirin/pharmacology , Cell Movement/drug effects , Endothelial Cells/drug effects , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Humans , Lymphatic Metastasis/pathology , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
12.
J Agric Food Chem ; 65(11): 2299-2306, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28248106

ABSTRACT

The spread of cancer cells to distant organs, in a process called metastasis, is the main factor that contributes to most death in cancer patients. Vanillin, the vanilla flavoring agent, has been shown to suppress metastasis in a mouse model. Here, we evaluated the antimetastatic potential of the food additive divanillin, the homodimer of vanillin, and their structurally related compounds, apocynin and diapocynin, in hepatocellular carcinoma cells. The Transwell invasion assay showed that the dimeric forms exhibited a potency higher than those of vanillin and apocynin in inhibiting invasion, with IC50 values of 23.3 ± 7.4 to 41.3 ± 4.2 µM for the dimers, which are 26-34-fold lower than IC50 values of vanillin and apocynin (p < 0.05). Both monomeric and dimeric forms target regulation of the invasion process by inhibiting phosphorylation of FAK and Akt. Molecular docking studies suggested that the dimers should bind more tightly than vanillin and apocynin to the Y397 pocket of the FAK FERM domain. Thus, the food additive divanillin has antimetastatic potential greater than that of the flavoring agent vanillin.


Subject(s)
Acetophenones/pharmacology , Benzaldehydes/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasm Metastasis/prevention & control , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Acetophenones/chemistry , Benzaldehydes/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Dimerization , Humans , Molecular Docking Simulation , Neoplasm Metastasis/physiopathology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/physiopathology
13.
Biochem Biophys Res Commun ; 466(3): 493-8, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26375501

ABSTRACT

Circulating tumor cells (CTCs) are metastasizing epithelial cancer cells that adapt to survive when floating in bloodstream during metastasis. This condition can be mimicked in vitro by using non-adherent cell culture. The chemosensitivity of CTCs appears to correlate with the response of metastatic cancer patients to therapy, but chemoresistance is also frequently observed in advanced stage cancer patients, who have never previously received chemotherapy. We hypothesize that adaptation of epithelial cancer cells to become floating CTCs could lead to development of chemoresistance. Here, we explore whether chemoresistance is induced in epithelial cancer cells when cultured under non-adherent conditions. Increased paclitaxel-specific resistance was observed in floating cells compared to attached cells in H460, MCF-7, and HepG2 human cancer cell lines, by 15.6-, 3.9-, and 2.6-fold increases in IC50 values, respectively. qRT-PCR analysis showed that a paclitaxel-resistant ß-tubulin isotype, ßIVa-tubulin, was the most up-regulated gene compared with other ß-tubulin isotypes in H460 floating cells, concomitant with elevated ERK activation. ERK inhibitor treatment could attenuate the up-regulation of ßIVa-tubulin, and decreased the paclitaxel resistance of H460 floating cells, even though other ß-tubulin isotypes were up-regulated when the ERK activation was blocked. In conclusion, we show induction of paclitaxel resistance in epithelial cancer cells, when floating in non-adherent culture, and this might occur with CTCs of cancer patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Paclitaxel/pharmacology , Tubulin/genetics , Cell Adhesion , Cell Culture Techniques , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Hep G2 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL