Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 238
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Formos Med Assoc ; 123(4): 442-451, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37805307

ABSTRACT

BACKGROUND: The study aimed to observe molecular signaling, including reactive oxygen species (ROS) and mitochondrial membrane potential (ΔΨm), to evaluate the alteration of gene expression by low-level laser therapy (LLLT) and the correlation between its mechanisms and the NF-kB pathway in cells involved in orthodontic tooth movement. METHODS: Osteoblast-like cells (MG63), immortalized periodontal ligament cells (iPDL), and M1 macrophage-like cells were irradiated by 980-nm LLLT with energy densities of 1 and 10 J/cm2 ΔΨm and intracellular ROS were monitored using fluorescent probes. The changes of mRNA expression were assessed using reverse transcription polymerase chain reaction (RT-PCR). NF-kB inhibitor, ROS scavenger, and ΔΨm suppressor were used to analyze signals associated with the regulation of gene expression. Finally, Western blot analysis was performed to confirm NF-kB signaling after LLLT. RESULTS: We found the increases of ΔΨm and ROS in all three cell types after LLLT, but no significant difference was observed between 1 and 10 J/cm2 LLLT. Regarding gene expression, some target genes were upregulated in MG63 6 h, 12 h, and 1 day after LLLT and in iPDL cells 12 h and 1 day after LLLT. However, no changes occurred in M1 cells. The inhibitor that significantly reduced most changes in gene expression was NF-kB inhibitor. Western blot analysis showed the increase in p-IkBα level after LLLT in iPDL and MG63, but not in M1. CONCLUSION: The 980-nm LLLT increased ΔΨm and ROS production in all three cell types. However, changes in gene regulation were found only in MG63 and iPDL cells, which related to the NF-kB pathway.


Subject(s)
NF-kappa B , Tooth Movement Techniques , Humans , Reactive Oxygen Species/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Lasers , Gene Expression
2.
J Biol Chem ; 298(6): 102036, 2022 06.
Article in English | MEDLINE | ID: mdl-35588785

ABSTRACT

Serum- and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase that plays important roles in the cellular stress response. While SGK1 has been reported to restrain inflammatory immune responses, the molecular mechanisms involved remain elusive, especially in oral bacteria-induced inflammatory milieu. Here, we found that SGK1 curtails Porphyromonas gingivalis-induced inflammatory responses through maintaining levels of tumor necrosis factor receptor-associated factor (TRAF) 3, thereby suppressing NF-κB signaling. Specifically, SGK1 inhibition significantly enhances production of proinflammatory cytokines, including tumor necrosis factor α, interleukin (IL)-6, IL-1ß, and IL-8 in P. gingivalis-stimulated innate immune cells. The results were confirmed with siRNA and LysM-Cre-mediated SGK1 KO mice. Moreover, SGK1 deletion robustly increased NF-κB activity and c-Jun expression but failed to alter the activation of mitogen-activated protein kinase signaling pathways. Further mechanistic data revealed that SGK1 deletion elevates TRAF2 phosphorylation, leading to TRAF3 degradation in a proteasome-dependent manner. Importantly, siRNA-mediated traf3 silencing or c-Jun overexpression mimics the effect of SGK1 inhibition on P. gingivalis-induced inflammatory cytokines and NF-κB activation. In addition, using a P. gingivalis infection-induced periodontal bone loss model, we found that SGK1 inhibition modulates TRAF3 and c-Jun expression, aggravates inflammatory responses in gingival tissues, and exacerbates alveolar bone loss. Altogether, we demonstrated for the first time that SGK1 acts as a rheostat to limit P. gingivalis-induced inflammatory immune responses and mapped out a novel SGK1-TRAF2/3-c-Jun-NF-κB signaling axis. These findings provide novel insights into the anti-inflammatory molecular mechanisms of SGK1 and suggest novel interventional targets to inflammatory diseases relevant beyond the oral cavity.


Subject(s)
Alveolar Bone Loss , Immediate-Early Proteins , Protein Serine-Threonine Kinases , TNF Receptor-Associated Factor 3 , Alveolar Bone Loss/genetics , Animals , Cytokines/metabolism , Genes, jun , Immediate-Early Proteins/metabolism , Immunity , Inflammation , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Porphyromonas gingivalis/pathogenicity , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/metabolism
3.
Am J Hum Genet ; 104(3): 422-438, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30773277

ABSTRACT

SPONASTRIME dysplasia is an autosomal-recessive spondyloepimetaphyseal dysplasia characterized by spine (spondylar) abnormalities, midface hypoplasia with a depressed nasal bridge, metaphyseal striations, and disproportionate short stature. Scoliosis, coxa vara, childhood cataracts, short dental roots, and hypogammaglobulinemia have also been reported in this disorder. Although an autosomal-recessive inheritance pattern has been hypothesized, pathogenic variants in a specific gene have not been discovered in individuals with SPONASTRIME dysplasia. Here, we identified bi-allelic variants in TONSL, which encodes the Tonsoku-like DNA repair protein, in nine subjects (from eight families) with SPONASTRIME dysplasia, and four subjects (from three families) with short stature of varied severity and spondylometaphyseal dysplasia with or without immunologic and hematologic abnormalities, but no definitive metaphyseal striations at diagnosis. The finding of early embryonic lethality in a Tonsl-/- murine model and the discovery of reduced length, spinal abnormalities, reduced numbers of neutrophils, and early lethality in a tonsl-/- zebrafish model both support the hypomorphic nature of the identified TONSL variants. Moreover, functional studies revealed increased amounts of spontaneous replication fork stalling and chromosomal aberrations, as well as fewer camptothecin (CPT)-induced RAD51 foci in subject-derived cell lines. Importantly, these cellular defects were rescued upon re-expression of wild-type (WT) TONSL; this rescue is consistent with the hypothesis that hypomorphic TONSL variants are pathogenic. Overall, our studies in humans, mice, zebrafish, and subject-derived cell lines confirm that pathogenic variants in TONSL impair DNA replication and homologous recombination-dependent repair processes, and they lead to a spectrum of skeletal dysplasia phenotypes with numerous extra-skeletal manifestations.


Subject(s)
Chromosomal Instability , DNA Damage , Genetic Variation , Musculoskeletal Abnormalities/pathology , NF-kappa B/genetics , Osteochondrodysplasias/pathology , Adolescent , Adult , Alleles , Animals , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Association Studies , Humans , Mice , Mice, Knockout , Musculoskeletal Abnormalities/genetics , Osteochondrodysplasias/genetics , Exome Sequencing , Young Adult , Zebrafish
4.
Ann Rheum Dis ; 81(10): 1453-1464, 2022 10.
Article in English | MEDLINE | ID: mdl-35868845

ABSTRACT

OBJECTIVES: To test the hypothesis that ROSAH (retinal dystrophy, optic nerve oedema, splenomegaly, anhidrosis and headache) syndrome, caused by dominant mutation in ALPK1, is an autoinflammatory disease. METHODS: This cohort study systematically evaluated 27 patients with ROSAH syndrome for inflammatory features and investigated the effect of ALPK1 mutations on immune signalling. Clinical, immunologic and radiographical examinations were performed, and 10 patients were empirically initiated on anticytokine therapy and monitored. Exome sequencing was used to identify a new pathogenic variant. Cytokine profiling, transcriptomics, immunoblotting and knock-in mice were used to assess the impact of ALPK1 mutations on protein function and immune signalling. RESULTS: The majority of the cohort carried the p.Thr237Met mutation but we also identified a new ROSAH-associated mutation, p.Tyr254Cys.Nearly all patients exhibited at least one feature consistent with inflammation including recurrent fever, headaches with meningeal enhancement and premature basal ganglia/brainstem mineralisation on MRI, deforming arthritis and AA amyloidosis. However, there was significant phenotypic variation, even within families and some adults lacked functional visual deficits. While anti-TNF and anti-IL-1 therapies suppressed systemic inflammation and improved quality of life, anti-IL-6 (tocilizumab) was the only anticytokine therapy that improved intraocular inflammation (two of two patients).Patients' primary samples and in vitro assays with mutated ALPK1 constructs showed immune activation with increased NF-κB signalling, STAT1 phosphorylation and interferon gene expression signature. Knock-in mice with the Alpk1 T237M mutation exhibited subclinical inflammation.Clinical features not conventionally attributed to inflammation were also common in the cohort and included short dental roots, enamel defects and decreased salivary flow. CONCLUSION: ROSAH syndrome is an autoinflammatory disease caused by gain-of-function mutations in ALPK1 and some features of disease are amenable to immunomodulatory therapy.


Subject(s)
Hereditary Autoinflammatory Diseases , NF-kappa B , Protein Kinases/genetics , Amyloidosis , Animals , Cohort Studies , Gain of Function Mutation , Hereditary Autoinflammatory Diseases/genetics , Humans , Inflammation/genetics , Mice , Mutation , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Kinases/metabolism , Quality of Life , Serum Amyloid A Protein , Syndrome , Tumor Necrosis Factor Inhibitors
5.
Eur J Orthod ; 44(6): 669-678, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35924725

ABSTRACT

OBJECTIVES: This study investigated the role of lncRNA growth arrest-specific transcript 5 (GAS5) in the inflammatory response of periodontal ligament stem cells (PDLSCs) during periodontitis with attempts to its possible mechanisms. MATERIALS AND METHODS: Gingiva samples were collected from healthy people and patients with periodontitis. The ligature-induced periodontitis model was established in mice. Cell transfection was utilized to knock down and overexpress GAS5 in PDLSCs. Quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization were performed to detect the GAS5 expression. In combination with high-throughput sequencing technology, qRT-PCR, Western blotting, and immunofluorescence were performed to detect the effects of GAS5 on cytokines and proteins in the NF-κB pathway. RESULTS: GAS5 expression decreased in PDLSCs subjected to compressive force. GAS5 expression was downregulated in the gingiva tissues from patients with periodontitis. Consistent with the results of clinical samples, GAS5 expression decreased in the mouse ligature-induced periodontitis model. GAS5 expression was downregulated in PDLSCs under tumour necrosis factor (TNF)-α stimulation. Knockdown and overexpression of GAS5 increased and decreased the expression of cytokines induced by TNF-α in PDLSCs, respectively. The sequencing results showed that overexpressing GAS5 was related to genes in the NF-κB pathway. Overexpressing GAS5 alleviated p65 phosphorylation and inhibited the entry of p65 into the nucleus in the TNF-α activated NF-κB pathway, whereas GAS5 knockdown resulted in contrasting results. CONCLUSIONS: GAS5 alleviated the expression of cytokines in PDLSCs by inhibiting activation of the TNF-α-mediated NF-κB pathway. These findings provide new insight into the regulation of the PDLSCs inflammation response.


Subject(s)
Periodontitis , RNA, Long Noncoding , Animals , Humans , Mice , Cell Differentiation , Cells, Cultured , In Situ Hybridization, Fluorescence , NF-kappa B/genetics , NF-kappa B/metabolism , Osteogenesis , Periodontal Ligament/metabolism , Periodontitis/genetics , Periodontitis/metabolism , Periodontitis/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Tumor Necrosis Factor-alpha/metabolism
6.
J Cell Mol Med ; 25(1): 333-344, 2021 01.
Article in English | MEDLINE | ID: mdl-33314684

ABSTRACT

Simvastatin (SMV) could increase tooth anchorage during orthodontic tooth movement (OTM). However, previous studies on its bone-specific anabolic and anti-inflammation properties were based on static in vitro and in vivo conditions. AMPK is a stress-activated kinase that protects tissue against serious damage from overloading inflammation. Rat periodontal ligament cells (PDLCs) were subjected to a serial of SMV concentrations to investigate the optimization that promoted osteogenic differentiation. The PDLCs in static and/or tensile culturing conditions then received the proper concentration SMV. Related factors expression was measured by the protein array, real-time PCR and Western blot. The 0.05UM SMV triggered osteogenic differentiation of PDLCs. The inhibition of AMPK activation through a pharmacological approach (Compound C) caused dramatic decrease in osteogenic/angiogenic gene expression and significant increase in inflammatory NF-κB phosphorylation. In contrast, pharmacological activation of AMPK by AICAR significantly inhibited inflammatory factors expression and activated ERK1/2, P38 MAPK phosphorylation. Moreover, AMPK activation induced by SMV delivery significantly attenuated the osteoclastogenesis and decreased the expression of pro-inflammatory TNF-α and NF-κB in a rodent model of OTM. The current studies suggested that SMV could intrigue intrinsic activation of AMPK in PDLCs that promote attenuate the inflammation which occurred under tensile irritation through AMPK/MAPK/NF-kB Inhibition.


Subject(s)
AMP-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Simvastatin/therapeutic use , AMP-Activated Protein Kinases/genetics , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Humans , NF-kappa B/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Mechanical , X-Ray Microtomography
7.
J Cell Mol Med ; 25(2): 1299-1313, 2021 01.
Article in English | MEDLINE | ID: mdl-33336563

ABSTRACT

The hepatic stellate cells (HSCs) play a significant role in the onset of liver fibrosis, which can be treated by the inhibition and reversal of HSC activation. The RNA interference-mediated TLR4 gene silencing might be a potential therapeutic approach for liver fibrosis. The crucial challenge in this method is the absence of an efficient delivery system for the RNAi introduction in the target cells. HSCs have an enhanced capacity of vitamin A intake as they contain retinoic acid receptors (RARs). In the current study, we developed cationic liposomes modified with vitamin A to improve the specificity of delivery vehicles for HSCs. The outcome of this study revealed that the VitA-coupled cationic liposomes delivered the TLR4 shRNA to aHSCs more efficiently, as compared to the uncoupled cationic liposomes, both in the in vitro and in vivo conditions. Besides, as evident from the outcome of this study, the TLR4 gene silencing inhibited the HSCs activation and attenuated the liver fibrosis via the NF-κB transcriptional inactivation, pro-inflammatory cytokines secretion and reactive oxygen species (ROS) synthesis. Thus, the VitA-coupled liposomes encapsulated with the TLR4-shRNA might prove as an efficient therapeutic agent for liver fibrosis.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/therapy , RNA, Small Interfering/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cytokines/metabolism , DNA/metabolism , Disease Progression , Extracellular Matrix/metabolism , Female , Gene Silencing , Inflammation Mediators/metabolism , Lipopolysaccharides , Liposomes , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Mitochondria/metabolism , NADPH Oxidases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Tissue Distribution , Transcription, Genetic , Vitamin A/metabolism , Vitamin A/pharmacokinetics
8.
Bioconjug Chem ; 32(8): 1729-1741, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34282895

ABSTRACT

Conjugation with poly(ethylene glycol) ("PEGylation") is a widely used approach for improving the therapeutic propensities of peptide and protein drugs through prolonging bloodstream circulation, reducing toxicity and immunogenicity, and improving proteolytic stability. In the present study, we investigate how PEGylation affects the interaction of host defense peptides (HDPs) with bacterial lipopolysaccharide (LPS) as well as HDP suppression of LPS-induced cell activation. In particular, we investigate the effects of PEGylation site for KYE28 (KYEITTIHNLFRKLTHRLFRRNFGYTLR), a peptide displaying potent anti-inflammatory effects, primarily provided by its N-terminal part. PEGylation was performed either in the N-terminus, the C-terminus, or in both termini, keeping the total number of ethylene groups (n = 48) constant. Ellipsometry showed KYE28 to exhibit pronounced affinity to both LPS and its hydrophobic lipid A moiety. The PEGylated peptide variants displayed lower, but comparable, affinity for both LPS and lipid A, irrespective of the PEGylation site. Furthermore, both KYE28 and its PEGylated variants triggered LPS aggregate disruption. To investigate the peptide structure in such LPS complexes, a battery of nuclear magnetic resonance (NMR) methods was employed. From this, it was found that KYE28 formed a well-folded structure after LPS binding, stabilized by hydrophobic domains involving aromatic amino acids as well as by electrostatic interactions. In contrast, the PEGylated peptide variants displayed a less well-defined secondary structure, suggesting weaker LPS interactions in line with the ellipsometry findings. Nevertheless, the N-terminal part of KYE28 retained helix formation after PEGylation, irrespective of the conjugation site. For THP1-Xblue-CD14 reporter cells, KYE28 displayed potent suppression of LPS activation at simultaneously low cell toxicity. Interestingly, the PEGylated KYE28 variants displayed similar or improved suppression of LPS-induced cell activation, implying the underlying key role of the largely retained helical structure close to the N-terminus, irrespective of PEGylation site. Taken together, the results show that PEGylation of HDPs can be done insensitively to the conjugation site without losing anti-inflammatory effects, even for peptides inducing such effects through one of its termini.


Subject(s)
Lipid A/chemistry , Lipopolysaccharides/chemistry , Peptides/chemistry , Polyethylene Glycols , Cell Line , Gene Expression Regulation/drug effects , Hemolysis , Humans , Models, Molecular , NF-kappa B/genetics , NF-kappa B/metabolism , Peptides/pharmacology , Protein Binding , Protein Conformation , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
9.
Ecotoxicol Environ Saf ; 227: 112882, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34700168

ABSTRACT

Microplastics (< 5 mm diameter) are one of most important environmental pollutants and contaminants worldwide. However, how microplastics affect liver immune microenvironment in not well understood. Microplastics (0.5 µm) were administered orally to C57BL/6J mice for 4 consecutive weeks at the rate of 0.5 mg/day. Non-parenchymal cells were isolated from of the mice through fractionation of fresh hepatic tissues. The immune landscape for four cell populations of B cells, T cells, NK cells and macrophages in the liver tissues was then evaluated using flow cytometry. The secretion level of inflammatory cytokines and associated signaling pathway were investigated using quantitative real-time polymerase chain reaction and western blot. Oral ingestion of microplastics increases liver weight, general liver index as well as expression of serum, liver function-related indicators. Microplastics also increased the infiltration of natural killer cells and macrophages to non-parenchymal liver cells, but reduced that of B cells to the same tissues. However, microplastics had no effect on the infiltration of T cell to non-parenchymal liver cells. Ingestion of MPs also up-regulated the expression of IFN-γ, TNF-α, IL-1ß, IL-6 and IL-33 mRNA, but down-regulated that of IL-4, IL-5, IL-10, IL-18 and TGF-ß1. Overall, the aforementioned processes were regulated via the NF-κB pathway in the hepatic non-parenchymal cells. Microplastics disrupts inflammatory process in liver tissues via the NF-κB signaling pathway. These findings provide a strong foundation on immune processes in hepatic tissues following prolonged ingestion of microplastics.


Subject(s)
Microplastics , Plastics , Animals , Eating , Inflammation/chemically induced , Killer Cells, Natural , Liver , Macrophages , Mice , Mice, Inbred C57BL , NF-kappa B/genetics
10.
J Cell Mol Med ; 24(5): 3053-3063, 2020 03.
Article in English | MEDLINE | ID: mdl-32022398

ABSTRACT

S100B protein bridges chronic mucosal inflammation and colorectal cancer given its ability to activate NF-kappaB transcription via RAGE signalling and sequestrate pro-apoptotic wtp53. Being an S100B inhibitor, pentamidine antagonizes S100B-wtp53 interaction, restoring wtp53-mediated pro-apoptotic control in cancer cells in several types of tumours. The expression of S100B, pro-inflammatory molecules and wtp53 protein was evaluated in human biopsies deriving from controls, ulcerative colitis and colon cancer patients at baseline (a) and (b) following S100B targeting with niosomal PENtamidine VEhiculation (PENVE), to maximize drug permeabilization in the tissue. Cultured biopsies underwent immunoblot, EMSA, ELISA and biochemical assays for S100B and related pro-inflammatory/pro-apoptotic proteins. Exogenous S100B (0.005-5 µmol/L) alone, or in the presence of PENVE (0.005-5 µmol/L), was tested in control biopsies while PENVE (5 µmol/L) was evaluated on control, peritumoral, ulcerative colitis and colon cancer biopsies. Our data show that S100B level progressively increases in control, peritumoral, ulcerative colitis and colon cancer enabling a pro-inflammatory/angiogenic and antiapoptotic environment, featured by iNOS, VEGF and IL-6 up-regulation and wtp53 and Bax inhibition. PENVE inhibited S100B activity, reducing its capability to activate RAGE/phosphor-p38 MAPK/NF-kappaB and favouring its disengagement with wtp53. PENVE blocks S100B activity and rescues wtp53 expression determining pro-apoptotic control in colon cancer, suggesting pentamidine as a potential anticancer drug.


Subject(s)
Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Pentamidine/administration & dosage , S100 Calcium Binding Protein beta Subunit/genetics , Tumor Suppressor Protein p53/genetics , Antigens, Neoplasm/genetics , Biopsy , Carcinoma/genetics , Carcinoma/pathology , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Liposomes/administration & dosage , Male , Middle Aged , Mitogen-Activated Protein Kinases/genetics , Mucous Membrane/drug effects , NF-kappa B/genetics , Tumor Microenvironment/drug effects
11.
J Cell Physiol ; 235(4): 3519-3528, 2020 04.
Article in English | MEDLINE | ID: mdl-31595494

ABSTRACT

Gestational diabetes mellitus (GDM) is an important factor involved in the pathogenesis of organ development in the offspring. Here, we analyzed the effects of GDM on odontoblastic differentiation of dental papilla cells (DPCs) and dentin formation in offspring and investigated their underlying mechanisms. A GDM rat model was induced by intraperitoneal injection of streptozotocin and offspring were collected. The results showed that GDM significantly affected odontoblast differentiation and dentin formation in offspring tooth. GDM activated the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-ĸB) signaling pathway and inhibited SMAD1/5/9 signaling to modulate the odontoblastic differentiation of DPCs in offspring. Inhibition of TLR4 signaling by treated with TAK-242 significantly reverses the suppression of odonto-differentiation of DPCs in diabetic offspring. Taken together, these data indicate GDM activated the offspring DPCs TLR4/NF-ĸB signaling, which suppressed the SMAD1/5/9 phosphorylation and then inhibited odontoblasts differentiation and dentin formation.


Subject(s)
Cell Differentiation/genetics , Dental Papilla/growth & development , Diabetes, Gestational/genetics , Toll-Like Receptor 4/genetics , Animals , Calcification, Physiologic/genetics , Cell Proliferation/drug effects , Dental Papilla/metabolism , Dental Pulp/growth & development , Dental Pulp/pathology , Diabetes, Gestational/pathology , Female , Humans , NF-kappa B/genetics , Odontoblasts/metabolism , Phosphorylation/genetics , Pregnancy , Rats , Signal Transduction/genetics , Smad1 Protein , Sulfonamides/pharmacology
12.
J Cell Physiol ; 235(12): 9691-9701, 2020 12.
Article in English | MEDLINE | ID: mdl-32399963

ABSTRACT

We wished to evaluate whether epigenetic modifiers have a beneficial effect on treating experimental periodontitis and mechanisms for regulating the cell fate of mesenchymal stem cells (MSCs) in inflammatory microenvironments. We isolated MSCs from healthy and inflamed gingival tissues to investigate whether trichostatin A (TSA) could improve osteogenic differentiation and resolve inflammation in vitro. The tissue regenerative potentials were evaluated when treated with a temperature-dependent, chitosan-scaffold-encapsulated TSA, in a rat model of periodontitis. After induction with the conditioned medium, TSA treatment increased the osteogenic differentiation potential of inflamed MSCs and healthy MSCs. In addition, interleukin-6 and interleukin-8 levels in supernatants were significantly decreased after TSA treatment. Moreover, TSA promoted osteogenic differentiation by inhibiting nuclear factor-κB (p65) DNA binding in MSCs. In rats with experimental periodontitis, 7 weeks after local injections of chitosan-scaffold-encapsulated TSA, histology and microcomputed tomography showed a significant increase in alveolar bone volume and less inflammatory infiltration compared with vehicle-treated rats. The concentrations of interferon-γ and interleukin-6 were significantly decreased in the gingival crevicular fluid after TSA treatment. This study demonstrated that TSA had anti-inflammatory properties and could promote periodontal tissue repair, which indicated that epigenetic modifiers hold promise as a potential therapeutic option for periodontal tissue repair.


Subject(s)
Cell Differentiation/drug effects , Hydroxamic Acids/pharmacology , Osteogenesis/genetics , Periodontium/growth & development , Animals , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , Disease Models, Animal , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , NF-kappa B/genetics , Osteogenesis/drug effects , Periodontium/diagnostic imaging , Periodontium/metabolism , Periodontium/pathology , Rats , X-Ray Microtomography
13.
Cancer Sci ; 111(4): 1344-1356, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31960547

ABSTRACT

5-Fluorouracil-based therapy remains the main approach in colorectal cancer, even though there are still some drawbacks, such as chemoresistance. In this study we combined 5-fluorouracil encapsulated in long-circulating liposomes with simvastatin, also encapsulated in long-circulating liposomes, that was previously proved to exert antitumor actions on the same tumor model. The production of angiogenic/inflammatory proteins was assessed by protein array and the production of markers for tumor aggressiveness (Bcl-2, Bax, and nuclear factor [NF]-κB) were determined by western blot analysis. Intratumor oxidative stress was evaluated through measurement of malondialdehyde level by HPLC, and through spectrophotometric analysis of catalytic activity of catalase and of total antioxidant capacity. Immunohistochemical analysis of tumors for CD31 expression was assessed. Intratumor activity of MMP-2 by gelatin zymography was also carried out. Our results revealed that combined therapies based on liposomal formulations exerted enhanced antitumor activities compared with combined treatment with free drugs. Sequential treatment with liposomal simvastatin and liposomal 5-fluorouracil showed the strongest antitumor activity in C26 colon carcinoma in vivo, mainly through inhibition of tumor angiogenesis. Important markers for cancer progression (Bcl-2, Bax, NF-κB, and intratumor antioxidants) showed that liposomal simvastatin might sensitize C26 cells to liposomal 5-fluorouracil treatment in both regimens tested. The outcome of simultaneous treatment with liposomal formulations was superior to sequential treatment with both liposomal types as the invasive capacity of C26 tumors was strongly increased after the latest treatment. The antitumor efficacy of combined therapy in C26 colon carcinoma might be linked to the restorative effects on proteins balance involved in tumor angiogenesis.


Subject(s)
Carcinoma/drug therapy , Colorectal Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Simvastatin/pharmacology , Animals , Apoptosis/drug effects , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liposomes/pharmacology , Mice , NF-kappa B/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics
14.
FASEB J ; 33(9): 10515-10527, 2019 09.
Article in English | MEDLINE | ID: mdl-31251083

ABSTRACT

Histone Lys-specific demethylases (KDMs) play a key role in many biological processes through epigenetic mechanisms. However, the role of KDMs in inflammatory responses to oral bacterial infection is poorly understood. Here, we show a novel regulatory role of KDM3C in inflammatory responses to oral bacterial infection. KDM3C expression is transiently suppressed in human and mouse macrophages exposed to LPS from Porphyromonas gingivalis (Pg LPS). Loss of KDM3C in both human and mouse macrophages led to notable induction of proinflammatory cytokines in response to Pg LPS stimulation. Also, KDM3C depletion led to strong induction of p65 phosphorylation and accelerated nuclear translocation in cells exposed to Pg LPS. Kdm3C knockout (KO) in mice led to increased alveolar bone destruction upon induction of experimental periodontitis or pulp exposure compared with those of the wild-type (WT) littermates. The Kdm3C KO mice also revealed an increased number of osteoclasts juxtaposed to the bony lesions. We also confirmed enhanced osteoclastogenesis by bone marrow-derived macrophages isolated from the Kdm3C KO compared with the WT controls. These findings suggest an anti-inflammatory function of KDM3C in regulating the inflammatory responses against oral bacterial infection through suppression of NF-κB signaling and osteoclastogenesis.-Lee, J. Y., Mehrazarin, S., Alshaikh, A., Kim, S., Chen, W., Lux, R., Gwack, Y., Kim, R. H., Kang, M. K. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-κB signaling and osteoclastogenesis.


Subject(s)
Inflammation/prevention & control , Jumonji Domain-Containing Histone Demethylases/physiology , Mouth Diseases/prevention & control , NF-kappa B/antagonists & inhibitors , Osteogenesis , Porphyromonas gingivalis/pathogenicity , Animals , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , Cell Differentiation , Cytokines , Histones , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Mouth Diseases/etiology , Mouth Diseases/metabolism , Mouth Diseases/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteoclasts/pathology , Phosphorylation , Signal Transduction
15.
Environ Toxicol ; 35(4): 495-506, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31797534

ABSTRACT

Microplastics (MPs) pollution is a global paradigm that raises concern in relation to environment and human health. In order to investigate the molecular toxicity mechanisms of MPs, transcriptomic analyses were performed on in vitro Caco-2 cell model. After observing that polystyrene microplastics (PS-MPs) decreased cell viability in a dose-dependent manner, the responsible genes and involved pathways that might make contribution to PS-MBs-induced toxicity to Caco-2 cells were identified with Illumina RNA seq. A total of 442 genes including, 210 up-regulated ones and 232 down-regulated ones, showed differential expression after treatment by PS-MPs with a concentration of 12.5 mg L-1 or 50.0 mg L-1 for 24 hours. Gene Ontology (GO) annotation enriched unigenes can be grouped into three separated clusters: cellular component (CC), biological process (BP), and molecular function (MF). The dominate pathways related to NF-κB, MAPK signaling, cytokine-cytokine receptor interaction, and toll-like receptor were strongly influenced by PS-MBs. These pathways are involved in modulating cell inflammatory and proliferation. The qPCR were applied to investigate the transcriptional level of five proliferation related genes (Ras, ERK, MER, CDK4, Cyclin D1) and four inflammation related genes (TRPV1, iNOS, IL-1ß, IL-8), and the results were consistent with RNA-seq data. This study has provided new insight into the understanding of the toxicity effects of PS-MBs-induced intestinal inflammatory diseases.


Subject(s)
Environmental Pollutants/toxicity , Microplastics/toxicity , Polystyrenes/toxicity , Transcriptome/drug effects , Caco-2 Cells , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Profiling , Gene Ontology , Humans , Inflammation , Interleukin-1beta/genetics , Microspheres , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Up-Regulation
16.
Int J Mol Sci ; 21(21)2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33142808

ABSTRACT

Polyethylene glycol (PEG) coating of gold nanoparticles (AuNPs) improves AuNP distribution via blood circulation. The use of PEG-coated AuNPs was shown to result in acute injuries to the liver, kidney, and spleen, but long-term toxicity has not been well studied. In this study, we investigated reporter induction for up to 90 days in NF-κB transgenic reporter mice following intravenous injection of PEG-coated AuNPs. The results of different doses (1 and 4 µg AuNPs per gram of body weight), particle sizes (13 nm and 30 nm), and PEG surfaces (methoxyl- or carboxymethyl-PEG 5 kDa) were compared. The data showed up to 7-fold NF-κB reporter induction in mouse liver from 3 h to 7 d post PEG-AuNP injection compared to saline-injected control mice, and gradual reduction to a level similar to control by 90 days. Agglomerates of PEG-AuNPs were detected in liver Kupffer cells, but neither gross pathological abnormality in liver sections nor increased activity of liver enzymes were found at 90 days. Injection of PEG-AuNPs led to an increase in collagen in liver sections and elevated total serum cholesterol, although still within the normal range, suggesting that inflammation resulted in mild fibrosis and affected hepatic function. Administrating PEG-AuNPs inevitably results in nanoparticles entrapped in the liver; thus, further investigation is required to fully assess the long-term impacts by PEG-AuNPs on liver health.


Subject(s)
Gold/chemistry , Inflammation/pathology , Liver/pathology , Metal Nanoparticles/toxicity , NF-kappa B/genetics , Polyethylene Glycols/chemistry , Animals , Inflammation/chemically induced , Inflammation/metabolism , Liver/drug effects , Liver/metabolism , Luciferases , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism
17.
Int J Mol Sci ; 21(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370039

ABSTRACT

It was hypothesized that strontium (Sr)-doped ß-tricalcium phosphate (TCP)-based scaffolds have a positive effect on the regeneration of large bone defects (LBD). Readouts in our mice models were nuclear factor-kappa beta (NF-κB) activity and vascular endothelial growth factor receptor-2 (VEGFR-2) promoter activity during the healing process. A 2-mm critical-size femoral fracture was performed in transgenic NF-κB- and VEGFR-2-luciferase reporter mice. The fracture was filled with a 3D-printed ß-TCP scaffold with or without Sr. A bioluminescence in-vivo imaging system was used to sequentially investigate NF-κB and VEGFR-2 expression for two months. After sacrifice, soft and osseous tissue formation in the fracture sites was histologically examined. NF-κB activity increased in the ß-TCP + Sr group in the latter stage (day 40-60). VEGFR-2 activity increased in the + Sr group from days 0-15 but decreased and showed significantly less activity than the ß-TCP and non-scaffold groups from days 40-60. The new bone formation and soft tissue formation in the + Sr group were significantly higher than in the ß-TCP group, whereas the percentage of osseous tissue formation in the ß-TCP group was significantly higher than in the ß-TCP + Sr group. We analyzed longitudinal VEGFR-2 promoter activity and NF-κB activity profiles, as respective agents of angiogenesis and inflammation, during LBD healing. The extended inflammation phase and eventually more rapid resorption of scaffold caused by the addition of strontium accelerates temporary bridging of the fracture gaps. This finding has the potential to inform an improved treatment strategy for patients who suffer from osteoporosis.


Subject(s)
Calcium Phosphates/chemistry , NF-kappa B/genetics , Phosphatidylethanolamines/chemistry , Promoter Regions, Genetic , Strontium/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Bone Regeneration , Bone Substitutes , Bone and Bones/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , NF-kappa B/metabolism , Tissue Scaffolds , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Molecules ; 25(20)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092290

ABSTRACT

Conventional treatments for chronic periodontitis are less effective in controlling inflammation and often relapse. Therefore, it is necessary to explore an immunomodulatory medication as an adjuvant. Ginsenoside Rb3 (Rb3), one of the most abundant active components of ginseng, has been found to possess anti-inflammatory and immunomodulatory properties. Here, we detected the anti-inflammatory effect of Rb3 on Porphyromonas gingivalis LPS-stimulated human periodontal ligament cells and experimental periodontitis rats for the first time. We found that the expression of pro-inflammatory mediators, including IL-1ß, IL-6 and IL-8, upregulated by lipopolysaccharide (LPS) stimulation was remarkably downregulated by Rb3 treatment in a dose-dependent manner at both transcriptional and translational levels. Network pharmacological analysis of Rb3 showed that the mitogen-activated protein kinase (MAPK) signaling pathway had the highest richness and that p38, JNK, and ERK molecules were potential targets of Rb3 in humans. Western blot analysis revealed that Rb3 significantly suppressed the phosphorylation of p38 MAPK and p65 NF-κB, as well as decreased the expression of total AKT. In experimental periodontitis rat models, reductions in alveolar bone resorption and osteoclast generation were observed in the Rb3 treatment group. Thus, we can conclude that Rb3 ameliorated Porphyromonas gingivalis LPS-induced inflammation by inhibiting the MAPK/AKT/NF-κB signaling pathways and attenuated alveolar bone resorption in experimental periodontitis rats.


Subject(s)
Alveolar Bone Loss/drug therapy , Cell Proliferation/drug effects , Ginsenosides/pharmacology , Inflammation/drug therapy , Alveolar Bone Loss/chemically induced , Alveolar Bone Loss/genetics , Alveolar Bone Loss/pathology , Animals , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/toxicity , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/genetics , Porphyromonas gingivalis/chemistry , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects
19.
J Cell Physiol ; 234(7): 11602-11609, 2019 07.
Article in English | MEDLINE | ID: mdl-30488444

ABSTRACT

DDIT3 is of great importance in endoplasmic reticulum stress and is involved in many inflammatory diseases and mineralization processes. The cementum protects teeth from periodontitis and provides attachment for Sharpey's fibers of the periodontal ligament. However, the effect of DDIT3 on cementoblast differentiation remains largely unknown. In this study, we found that DDIT3 was suppressed during cementoblast differentiation. Knockdown of DDIT3 increased the messenger RNA (mRNA) and protein levels of several key osteogenic markers in vitro, including alkaline phosphatase, runt-related transcription factor 2, and osteocalcin (OCN). In addition, isocitrate dehydrogenase 1 (IDH1) was increased during cementoblast differentiation, and knockdown of DDIT3 increased the protein and mRNA levels of IDH1. Furthermore, inhibition of IDH1 could partially reduce the effect of DDIT3 on cementoblast differentiation. The DDIT3 knockdown activated nuclear factor-κB (NF-κB) transcriptional activity and upregulated the expression of p-p65 and p-IκBα. The increased osteogenic differentiation ability and IDH1 expression, as induced by the DDIT3 knockdown, could be partially turned over by the addition of NF-κB inhibitor BAY 11-7082. Overall, our data clarified that DDIT3 suppresses cementoblast differentiation through IDH1, via the NF-κB pathway.


Subject(s)
Dental Cementum/metabolism , Isocitrate Dehydrogenase/metabolism , NF-kappa B/metabolism , Transcription Factor CHOP/metabolism , Animals , Cell Differentiation , Cell Line , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Isocitrate Dehydrogenase/genetics , Mice , NF-kappa B/genetics , Nitriles/pharmacology , Sulfones/pharmacology , Transcription Factor CHOP/genetics
20.
Exp Cell Res ; 372(2): 178-187, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30287143

ABSTRACT

Oral diseases, such as periapical periodontitis and periodontitis, are characterized by inflammation-induced bone loss. LL-37, a human antimicrobial peptide (AMP), has multiple biological functions and the potential to promote osteogenesis. Therefore, this study aimed to investigate the regulatory effects of LL-37 within normal and inflammatory microenvironments. The roles of P2X7 receptor (P2X7R) and mitogen-activated protein kinase (MAPK) signaling pathway were also demonstrated. The results showed that LL-37 promoted bone marrow stromal cell (BMSC) proliferation, migration and osteogenic differentiation. LL-37 inhibited the expression of the inflammatory cytokines interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and receptor activator of nuclear factor kappa-B ligand (RANKL) at both protein and gene levels, and attenuated the lipopolysaccharide (LPS)-induced inhibition of osteogenesis. Immunofluorescence (IF) confirmed P2X7R expression in BMSCs. BBG, a P2X7R antagonist, significantly attenuated LL-37-promoted osteogenesis. The phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH2-terminal kinase (JNK) increased after LL-37 stimulation, which did not affect p38 phosphorylation. The effects of LL-37 on osteogenesis-related gene expression were markedly attenuated by selective inhibitors of ERK1/2 and JNK. Furthermore, a mouse model of LPS-stimulated calvarial osteolysis was established, and results showed that LL-37 markedly inhibited osteoclastic bone resorption. In conclusion, we speculate that LL-37 inhibits inflammation and promotes BMSC osteogenesis via P2X7R and MAPK signaling pathway.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Inflammation/drug therapy , Mesenchymal Stem Cells/drug effects , Osteogenesis/genetics , Receptors, Purinergic P2X7/drug effects , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/genetics , Alveolar Bone Loss/pathology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cellular Microenvironment/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/genetics , Osteogenesis/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL