Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Inflamm Res ; 72(4): 769-782, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36813915

ABSTRACT

BACKGROUND: The mechanism by which incompletely absorbed fructose causes gastrointestinal symptoms is not fully understood. In this study, we investigated the immunological mechanisms of bowel habit changes associated with fructose malabsorption by examining Chrebp-knockout mice exhibiting defective fructose absorption. METHODS: Mice were fed a high-fructose diet (HFrD), and stool parameters were monitored. The gene expression in the small intestine was analyzed by RNA sequencing. Intestinal immune responses were assessed. The microbiota composition was determined by 16S rRNA profiling. Antibiotics were used to assess the relevance of microbes for HFrD-induced bowel habit changes. RESULTS: Chrebp-knockout (KO) mice fed HFrD showed diarrhea. Small-intestine samples from HFrD-fed Chrebp-KO mice revealed differentially expressed genes involved in the immune pathways, including IgA production. The number of IgA-producing cells in the small intestine decreased in HFrD-fed Chrebp-KO mice. These mice showed signs of increased intestinal permeability. Chrebp-KO mice fed a control diet showed intestinal bacterial imbalance, which the HFrD exaggerated. Bacterial reduction improved diarrhea-associated stool parameters and restored the decreased IgA synthesis induced in HFrD-fed Chrebp-KO mice. CONCLUSIONS: The collective data indicate that gut microbiome imbalance and disrupting homeostatic intestinal immune responses account for the development of gastrointestinal symptoms induced by fructose malabsorption.


Subject(s)
Diarrhea , Fructose , Mice , Animals , RNA, Ribosomal, 16S , Diarrhea/etiology , Transcription Factors/genetics , Transcription Factors/metabolism , Intestine, Small , Habits , Immunoglobulin A
2.
Biol Res ; 56(1): 40, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37438821

ABSTRACT

BACKGROUND: Polar microalgae contain unique compounds that enable them to adapt to extreme environments. As the skin barrier is our first line of defense against external threats, polar microalgae extracts may possess restorative properties for damaged skin, but the potential of microalgae extracts as skin protective agents remains unknown. PURPOSE: This study aimed to analyze compound profiles from polar microalgae extracts, evaluate their potential as skin epithelial protective agents, and examine the underlying mechanisms. METHODS: Six different polar microalgae, Micractinium sp. (KSF0015 and KSF0041), Chlamydomonas sp. (KNM0029C, KSF0037, and KSF0134), and Chlorococcum sp. (KSF0003), were collected from the Antarctic or Arctic regions. Compound profiles of polar and non-polar microalgae extracts were analyzed using gas chromatography-mass spectrometry (GC-MS). The protective activities of polar microalgae extracts on human keratinocyte cell lines against oxidative stress, radiation, and psoriatic cytokine exposure were assessed. The potential anti-inflammatory mechanisms mediated by KSF0041, a polar microalga with protective properties against oxidative stress, ultraviolet (UV) B, and an inflammatory cytokine cocktail, were investigated using RNA-sequencing analysis. To evaluate the therapeutic activity of KSF0041, an imiquimod-induced murine model of psoriatic dermatitis was used. RESULTS: Polar microalgae contain components comparable to those of their non-polar counterparts, but also showed distinct differences, particularly in fatty acid composition. Polar microalgae extracts had a greater ability to scavenge free radicals than did non-polar microalgae and enhanced the viability of HaCaT cells, a human keratinocyte cell line, following exposure to UVB radiation or psoriatic cytokines. These extracts also reduced barrier integrity damage and decreased mRNA levels of inflammatory cytokines in psoriatic HaCaT cells. Treatment with KSF0041 extract altered the transcriptome of psoriatic HaCaT cells toward a more normal state. Furthermore, KSF0041 extract had a therapeutic effect in a mouse model of psoriasis. CONCLUSIONS: Bioactive compounds from polar microalgae extracts could provide novel therapeutics for damaged and/or inflamed skin.


Subject(s)
Dermatitis , Microalgae , Humans , Animals , Mice , Keratinocytes , Cytokines , Protective Agents , Inflammation , Plant Extracts/pharmacology
3.
Vet Med (Praha) ; 67(9): 487-491, 2022 Jun.
Article in English | MEDLINE | ID: mdl-38715970

ABSTRACT

An 11-year-old intact female Pomeranian dog was referred for jaundice, anorexia, and vomiting. The blood analysis revealed increased alanine aminotransferase, alkaline phosphatase, and gamma-glutamyl transpeptidase. The serum canine pancreatic lipase immunoreactivity was within the normal reference range. The radiography revealed no significant findings. On ultrasound, the gallbladder was enlarged with a markedly distended common bile duct (CBD) measuring up to 6 mm in diameter. The pancreas had an irregular contour, a hypoechoic peripheral rim, multiple hyperechoic foci with acoustic shadowing, and showed increased echogenicity of the adjacent mesentery. Based on these results, an extrahepatic biliary obstruction secondary to the presumed chronic pancreatitis was diagnosed. The computed tomography (CT) images showed a hypoattenuating pancreatic parenchyma compared to the liver in the early phase, as well as multiple calcifications. A laparotomy was performed to reserve the patency of the CBD. The histopathological examination of the pancreas revealed exocrine pancreatic adenocarcinoma. While various appearances of exocrine pancreatic adenocarcinoma on CT have been reported in humans, CT features of pancreatic adenocarcinoma have not been well-established in dogs. The purpose of this report is to describe the atypical imaging features of pancreatic adenocarcinoma that are similar to those of chronic pancreatitis in a dog.

5.
Biochem Biophys Res Commun ; 463(3): 322-8, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26028561

ABSTRACT

An antimicrobial peptide (AMP), Hn-Mc, was designed by combining the N-terminus of HPA3NT3 and the C-terminus of melittin. This chimeric AMP exhibited potent antibacterial activity with low minimal inhibitory concentrations (MICs), ranging from 1 to 2 µM against four drug-susceptible bacteria and ten drug-resistant bacteria. Moreover, the hemolysis and cytotoxicity was reduced significantly compared to those of the parent peptides, highlighting its high cell selectivity. The morphological changes in the giant unilamellar vesicles and bacterial cell surfaces caused by the Hn-Mc peptide suggested that it killed the microbial cells by damaging the membrane envelope. An in vivo study also demonstrated the antibacterial activity of the Hn-Mc peptide in a mouse model infected with drug-resistant bacteria. In addition, the chimeric peptide inhibited the expression of lipopolysaccharide (LPS)-induced cytokines in RAW 264.7 cells by preventing the interaction between LPS and Toll-like receptors. These results suggest that this chimeric peptide is an antimicrobial and anti-inflammatory candidate as a pharmaceutic agent.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Melitten/chemistry , Peptide Fragments/chemistry , Ribosomal Proteins/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bacteria/drug effects , Bacteria/immunology , Bacterial Infections/drug therapy , Cell Line , Drug Resistance, Bacterial , Hemolysis/drug effects , Lipopolysaccharides/immunology , Melitten/chemical synthesis , Melitten/pharmacology , Melitten/therapeutic use , Mice , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Ribosomal Proteins/chemical synthesis , Ribosomal Proteins/pharmacology , Ribosomal Proteins/therapeutic use
6.
Mol Aspects Med ; 99: 101306, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191143

ABSTRACT

Psoriasis is a chronic inflammatory skin disease occurring worldwide. Initially viewed as a keratinocyte disorder, psoriasis is now recognized to involve a complex interplay between genetic predisposition, environmental triggers, and a dysregulated immune system, with a significant role of CD4+ T cells producing IL-17. Recent genetic studies have identified susceptibility loci that underscore the importance of innate immune responses, particularly the roles of myeloid cells, such as dendritic cells, macrophages, and neutrophils. These cells initiate and sustain inflammation through cytokine production triggered by external stimuli. They influence keratinocyte behavior and interact with adaptive immune cells. Recent techniques have further revealed the heterogeneity of myeloid cells in psoriatic lesions, highlighting the contributions of less-studied subsets, such as eosinophils and mast cells. This review examines the multifaceted roles of myeloid innate immune cells in psoriasis, emphasizing their functional diversity in promoting psoriatic inflammation. It also describes current treatment targeting myeloid innate immune cells and explores potential new therapeutic strategies based on the functional characteristics of these subsets. Future research should focus on the detailed characterization of myeloid subsets and their interactions to develop targeted treatments that address the complex immune landscape of psoriasis.


Subject(s)
Immunity, Innate , Myeloid Cells , Psoriasis , Humans , Psoriasis/immunology , Psoriasis/etiology , Psoriasis/genetics , Psoriasis/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , Dendritic Cells/immunology , Macrophages/immunology , Macrophages/metabolism , Neutrophils/immunology , Neutrophils/metabolism
7.
Exp Mol Med ; 56(5): 1164-1177, 2024 May.
Article in English | MEDLINE | ID: mdl-38689088

ABSTRACT

Recent evidence of gut microbiota dysbiosis in the context of psoriasis and the increased cooccurrence of inflammatory bowel disease and psoriasis suggest a close relationship between skin and gut immune responses. Using a mouse model of psoriasis induced by the Toll-like receptor (TLR) 7 ligand imiquimod, we found that psoriatic dermatitis was accompanied by inflammatory changes in the small intestine associated with eosinophil degranulation, which impaired intestinal barrier integrity. Inflammatory responses in the skin and small intestine were increased in mice prone to eosinophil degranulation. Caco-2 human intestinal epithelial cells were treated with media containing eosinophil granule proteins and exhibited signs of inflammation and damage. Imiquimod-induced skin and intestinal changes were attenuated in eosinophil-deficient mice, and this attenuation was counteracted by the transfer of eosinophils. Imiquimod levels and the distribution of eosinophils were positively correlated in the intestine. TLR7-deficient mice did not exhibit intestinal eosinophil degranulation but did exhibit attenuated inflammation in the skin and small intestine following imiquimod administration. These results suggest that TLR7-dependent bidirectional skin-to-gut communication occurs in psoriatic inflammation and that inflammatory changes in the intestine can accelerate psoriasis.


Subject(s)
Cell Degranulation , Disease Models, Animal , Eosinophils , Intestine, Small , Psoriasis , Toll-Like Receptor 7 , Animals , Humans , Mice , Caco-2 Cells , Eosinophils/metabolism , Eosinophils/immunology , Imiquimod , Inflammation/pathology , Inflammation/metabolism , Intestine, Small/pathology , Intestine, Small/metabolism , Membrane Glycoproteins , Mice, Knockout , Psoriasis/pathology , Psoriasis/metabolism , Skin/pathology , Skin/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics
8.
Gut Liver ; 17(1): 130-138, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36472070

ABSTRACT

Background/Aims: There are no data regarding the association between sarcopenic obesity status and nonalcoholic fatty liver disease (NAFLD) and NAFLD-associated liver fibrosis. Therefore, we aimed to investigate the relationship between sarcopenic obesity status (sarcopenia only, obesity only, and sarcopenic obesity) and NAFLD and liver fibrosis in Korean adults. Methods: In total, 2,191 subjects completed a health checkup program, including abdominal ultrasonography and FibroScan. Subjects were classified into the following four categories: optimal body composition (nonobese and nonsarcopenic), sarcopenia only (nonobese), obesity only (nonsarcopenic), and sarcopenic obesity. Sarcopenic obesity was stratified by the skeletal muscle mass index and body fat using bioelectrical impedance analysis. NAFLD was diagnosed by ultrasonography, and liver fibrosis was assessed using transient elastography in subjects with NAFLD. Results: The prevalence of NAFLD and liver fibrosis significantly increased according to the sarcopenic obesity status. In the logistic regression analysis, after adjusting for multiple risk factors, the odds ratio (OR) for the risk of NAFLD was largest in the sarcopenic obesity group (OR, 3.68; 95% confidence interval [CI], 2.94 to 4.60), followed by the obesity only (OR, 2.25; 95% CI, 1.67 to 3.03) and sarcopenia only (OR, 1.92; 95% CI, 1.30 to 2.84) groups, when compared with the optimal group. Additionally, liver fibrosis was independently associated with sarcopenic obesity status (OR 4.69, 95% CI 1.95 to 11.29; OR 4.17, 95% CI 1.56 to 11.17; OR 3.80, 95% CI 0.86 to 16.75, respectively). Conclusions: These results demonstrated that sarcopenic obesity was independently associated with NAFLD and liver fibrosis and increased the risk of NAFLD and liver fibrosis more than obesity or sarcopenia alone.


Subject(s)
Non-alcoholic Fatty Liver Disease , Sarcopenia , Adult , Humans , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/epidemiology , Sarcopenia/complications , Sarcopenia/epidemiology , Obesity/complications , Obesity/epidemiology , Fibrosis , Liver Cirrhosis/complications , Liver Cirrhosis/epidemiology , Liver Cirrhosis/diagnosis
9.
Cell Death Dis ; 12(3): 243, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664229

ABSTRACT

Atopic dermatitis is a chronic skin inflammatory disease mediated by Th2-type immune responses. Although intestinal immune responses have been shown to play a critical role in the development or prevention of atopic dermatitis, the precise influence of intestinal immunity on atopic dermatitis is incompletely understood. We show here that orally tolerized mice are protected from experimental atopic dermatitis induced by sensitization and epicutaneous (EC) challenge to ovalbumin. Although the expression of Th2-type cytokines in the small intestine of orally tolerized and EC-challenged mice did not change significantly, these mice showed decreased inflammatory responses in the small intestine with restoration of microbial change elicited by the EC challenge. Interestingly, an increase in small intestinal eosinophils was observed with the EC challenge, which was also inhibited by oral tolerance. The role of small intestinal eosinophils and microbiota in the pathogenesis of experimental atopic dermatitis was further substantiated by decreased inflammatory mediators in the small intestine and attenuated Th2-type inflammation in the skin of eosinophil-deficient and microbiota-ablated mice with EC challenges. Based on these data, we propose that the bidirectional interaction between the skin and the intestine has a role in the pathogenesis of atopic dermatitis and that modulation of the intestinal microenvironments could be a therapeutic approach to atopic dermatitis.


Subject(s)
Dermatitis, Atopic/prevention & control , Desensitization, Immunologic , Immune Tolerance , Intestine, Small/immunology , Leukocytes/immunology , Ovalbumin/administration & dosage , Skin/immunology , Administration, Oral , Animals , Bacteria/immunology , Claudin-4/genetics , Claudin-4/metabolism , Cytokines/genetics , Cytokines/metabolism , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/microbiology , Disease Models, Animal , Dysbiosis , Female , Gastrointestinal Microbiome , Host-Pathogen Interactions , Intestine, Small/metabolism , Intestine, Small/microbiology , Leukocytes/metabolism , Mice, Inbred BALB C , Skin/metabolism
11.
Front Cell Dev Biol ; 7: 329, 2019.
Article in English | MEDLINE | ID: mdl-31921842

ABSTRACT

Eosinophils are terminally differentiated granulocytes that have long been considered as destructive cells associated with Th2 type immune responses such as allergic inflammation and helminth infections. Recently, eosinophils have been actively studied as multifunctional leukocytes regulating an array of physiological responses through interaction with other immune cells. In this study, we examined the expression and function of Toll-like receptors (TLRs) in eosinophilic EoL-1 cells and demonstrated the expression of a number of immune mediators in activated EoL-1 cells and their interaction with the macrophage cell line THP-1 upon TLR4 ligand stimulation. EoL-1 cells differentiated with butyrate increased expression of TLR3, TLR4, and TLR7 at mRNA and protein level with flow cytometry analysis. Mature eosinophils derived from human cord blood CD34+ cells were subjected to RNA-sequencing, and showed the expression of a panel of TLR transcripts and TLR4 was the most highly expressed TLR. Among the cognate ligands of TLR3, TLR4, and TLR7, lipopolysaccharide (LPS) or palmitic acid significantly increased mRNA expression of immune mediators in differentiated EoL-1 cells. Notably, Western blot analysis of palmitic acid-treated differentiated EoL-1 cells showed significantly up-regulated expression of Th2 type cytokines and transcription factors driving eosinophil differentiation. To evaluate functional significance of TLR4 ligand-stimulated eosinophils, we added conditioned media (CM) from EoL-1 cells to differentiated THP-1 cells and assessed the expression of M1 macrophage or M2 macrophage-related markers. M1 and M2 macrophage markers were significantly upregulated by CM from LPS and palmitic acid stimulated EoL-1 cells, respectively. In addition, the adipose tissue of obese mice, where eosinophils are decreased due to obesity-induced inflammation, showed significantly decreased frequency of M2 macrophages, despite an increase in the total macrophage numbers. Based on these collective data, we proposed that eosinophils regulate both inflammatory and anti-inflammatory polarization of macrophages through functional changes induced by different TLR4 ligands.

12.
Sci Rep ; 8(1): 9894, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29967467

ABSTRACT

Accumulating data have indicated a fundamental role of eosinophils in regulating adipose tissue homeostasis. Here, we performed whole-genome RNA sequencing of the small intestinal tract, which suggested the presence of impaired lipid metabolism in eosinophil-deficient ΔdblGATA mice. ΔdblGATA mice fed a high-fat diet (HFD) showed reduced body fat mass, impaired enlargement of adipocytes, decreased expression of adipogenic genes, and developed glucose intolerance. HFD induced accumulation of eosinophils in the perigonadal white adipose tissue. Concordantly, adipocyte-differentiated 3T3-L1 cells promoted the migration of eosinophils through the expression of CCL11 (eotaxin-1) and likely promoted their survival through the expression of interleukin (IL)-3, IL-5, and granulocyte-macrophage colony-stimulating factor. HFD-fed ΔdblGATA mice showed increased infiltration of macrophages, CD4+ T-cells, and B-cells, increased expression of interferon-γ, and decreased expression of IL-4 and IL-13 in white adipose tissue. Interferon-γ treatment significantly decreased lipid deposition in adipocyte-differentiated 3T3-L1 cells, while IL-4 treatment promoted lipid accumulation. Notably, HFD-fed ΔdblGATA mice showed increased lipid storage in the liver as compared with wild-type mice. We propose that obesity promotes the infiltration of eosinophils into adipose tissue that subsequently contribute to the metabolic homeostasis by promoting adipocyte maturation.


Subject(s)
Adipocytes/pathology , Eosinophils/pathology , Obesity/pathology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/pathology , Animals , Cell Differentiation , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Cytokines/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Eosinophils/metabolism , GATA Transcription Factors/genetics , Glucose Tolerance Test , Interferon-gamma/pharmacology , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Obesity/etiology , Obesity/metabolism
13.
Cell Signal ; 28(8): 788-97, 2016 08.
Article in English | MEDLINE | ID: mdl-27049873

ABSTRACT

Bortezomib is an anti-cancer agent that induces ER stress by inhibiting proteasomal degradation. However, the effects of bortezomib appear to be dependent on its concentration and cellular context. Since ER stress is closely related to type 2 diabetes, the authors examined the effects of bortezomib on palmitic acid (PA)-induced ER stress in C2C12 murine myotubes. At low concentrations (<20nM), bortezomib protected myotubes from PA (750µM)-induced ER stress and inflammation. Either tunicamycin or thapsigargin-induced ER stress was also reduced by bortezomib. In addition, reduced glucose uptake and Akt phosphorylation induced by PA were prevented by co-treating bortezomib (10nM) both in the presence or absence of insulin. These protective effects of bortezomib were found to be associated with reduced JNK phosphorylation. Furthermore, bortezomib-induced AMPK phosphorylation, and the protective effects of bortezomib were diminished by AMPK knockdown, suggesting that AMPK activation underlies the effects of bortezomib. The in vivo administration of bortezomib at nontoxic levels (at 50 or 200µg/kg, i.p.) twice weekly for 5weeks to ob/ob mice improved insulin resistance, increased AMPK phosphorylation, reduced ER stress marker levels, and JNK inhibition in skeletal muscle. The study shows that bortezomib reduces ER stress, inflammation, and insulin resistance in vitro and in vivo, and suggests that bortezomib has novel applications for the treatment of metabolic disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bortezomib/pharmacology , Endoplasmic Reticulum Stress/drug effects , Inflammation/pathology , Insulin Resistance , Muscle Fibers, Skeletal/enzymology , Palmitic Acid/toxicity , Animals , Cell Line , Cytoprotection/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Male , Mice, Obese , Models, Biological , Muscle Fibers, Skeletal/drug effects , Thapsigargin/pharmacology , Tunicamycin/pharmacology
14.
PLoS One ; 11(5): e0155432, 2016.
Article in English | MEDLINE | ID: mdl-27176632

ABSTRACT

Lysimachia foenum-graecum extract (LFE) and its active component foenumoside B (FSB) have been shown to inhibit adipocyte differentiation, but their mechanisms were poorly defined. Here, we investigated the molecular mechanisms responsible for their anti-adipogenic effects. Both LFE and FSB inhibited the differentiation of 3T3-L1 preadipocytes induced by peroxisome proliferator-activated receptor-γ (PPARγ) agonists, accompanied by reductions in the expressions of the lipogenic genes aP2, CD36, and FAS. Moreover, LFE and FSB inhibited PPARγ transactivation activity with IC50s of 22.5 µg/ml and 7.63 µg/ml, respectively, and showed selectivity against PPARα and PPARδ. Rosiglitazone-induced interaction between PPARγ ligand binding domain (LBD) and coactivator SRC-1 was blocked by LFE or FSB, whereas reduced NCoR-1 binding to PPARγ by rosiglitazone was reversed in the presence of LFE or FSB. In vivo administration of LFE into either ob/ob mice or KKAy mice reduced body weights, and levels of PPARγ and C/EBPα in fat tissues. Furthermore, insulin resistance was ameliorated by LFE treatment, with reduced adipose tissue inflammation and hepatic steatosis. Thus, LFE and FSB were found to act as PPARγ antagonists that improve insulin sensitivity and metabolic profiles. We propose that LFE and its active component FSB offer a new therapeutic strategy for metabolic disorders including obesity and insulin resistance.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , PPAR gamma/antagonists & inhibitors , Primulaceae/chemistry , Saponins/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Carrier Proteins , Cell Line , Humans , Male , Mice , Models, Molecular , Molecular Conformation , PPAR gamma/chemistry , PPAR gamma/metabolism , Protein Binding , Saponins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL