Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.720
Filter
1.
J Cachexia Sarcopenia Muscle ; 13(1): 758-771, 2022 02.
Article in English | MEDLINE | ID: mdl-34997830

ABSTRACT

BACKGROUND: Chronic renal failure induces bone mineral disorders and sarcopenia. Skeletal muscle affects other tissues, including bone, by releasing myokines. However, the effects of chronic renal failure on the interactions between muscle and bone remain unclear. METHODS: We investigated the effects of renal failure on bone, muscle, and myokines linking muscle to bone using a mouse 5/6 nephrectomy (Nx) model. Muscle mass and bone mineral density (BMD) were analysed by quantitative computed tomography 8 weeks after Nx. RESULTS: Nephrectomy significantly reduced muscle mass in the whole body (12.1% reduction, P < 0.05), grip strength (10.1% reduction, P < 0.05), and cortical BMD at the femurs of mice (9.5% reduction, P < 0.01) 8 weeks after surgery, but did not affect trabecular BMD at the femurs. Among the myokines linking muscle to bone, Nx reduced the expression of irisin, a proteolytic product of fibronectin type III domain-containing 5 (Fndc5), in the gastrocnemius muscles of mice (38% reduction, P < 0.01). Nx increased myostatin mRNA levels in the gastrocnemius muscles of mice (54% increase, P < 0.01). In simple regression analyses, cortical BMD, but not trabecular BMD, at the femurs was positively related to Fndc5 mRNA levels in the gastrocnemius muscles of mice (r = 0.651, P < 0.05). The weekly administration of recombinant irisin to mice ameliorated the decrease in cortical BMD, but not muscle mass or grip strength, induced by Nx (6.2% reduction in mice with Nx vs. 3.3% reduction in mice with Nx and irisin treatment, P < 0.05). CONCLUSIONS: The present results demonstrated that renal failure decreases the expression of irisin in the gastrocnemius muscles of mice. Irisin may contribute to cortical bone loss induced by renal failure in mice as a myokine linking muscle to bone.


Subject(s)
Fibronectins , Renal Insufficiency , Animals , Bone and Bones/metabolism , Cortical Bone/metabolism , Fibronectins/biosynthesis , Fibronectins/genetics , Fibronectins/metabolism , Mice , Muscle, Skeletal/metabolism , Renal Insufficiency/metabolism
2.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884812

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) recapitulates metastasis and can be induced in vitro through transforming growth factor (TGF)-ß signaling. A role for MMP activity in glioblastoma multiforme has been ascribed to EMT, but the molecular crosstalk between TGF-ß signaling and membrane type 1 MMP (MT1-MMP) remains poorly understood. Here, the expression of common EMT biomarkers, induced through TGF-ß and the MT1-MMP inducer concanavalin A (ConA), was explored using RNA-seq analysis and differential gene arrays in human U87 glioblastoma cells. TGF-ß triggered SNAIL and fibronectin expressions in 2D-adherent and 3D-spheroid U87 glioblastoma cell models. Those inductions were antagonized by the TGF-ß receptor kinase inhibitor galunisertib, the JAK/STAT inhibitors AG490 and tofacitinib, and by the diet-derived epigallocatechin gallate (EGCG). Transient gene silencing of MT1-MMP prevented the induction of SNAIL by ConA and abrogated TGF-ß-induced cell chemotaxis. Moreover, ConA induced STAT3 and Src phosphorylation, suggesting these pathways to be involved in the MT1-MMP-mediated signaling axis that led to SNAIL induction. Our findings highlight a new signaling axis linking MT1-MMP to TGF-ß-mediated EMT-like induction in glioblastoma cells, the process of which can be prevented by the diet-derived EGCG.


Subject(s)
Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Glioblastoma/pathology , Matrix Metalloproteinase 14/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Snail Family Transcription Factors/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Concanavalin A , Fibronectins/biosynthesis , Humans , Matrix Metalloproteinase 14/genetics , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism , Tyrphostins/pharmacology
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298955

ABSTRACT

PURPOSE: The effects of Rho-associated coiled-coil containing protein kinase (ROCK) 1 and 2 inhibitor, ripasudil hydrochloride hydrate (Rip), ROCK2 inhibitor, KD025 or rosiglitazone (Rosi) on two-dimension (2D) and three-dimension (3D) cultured human conjunctival fibroblasts (HconF) treated by transforming growth factor (TGFß2) were studied. METHODS: Two-dimension and three-dimension cultured HconF were examined by transendothelial electrical resistance (TEER, 2D), size and stiffness (3D), and the expression of the extracellular matrix (ECM) including collagen1 (COL1), COL4 and COL6, fibronectin (FN), and α-smooth muscle actin (αSMA) by quantitative PCR (2D, 3D) in the presence of Rip, KD025 or Rosi. RESULTS: TGFß2 caused a significant increase in (1) the TEER values (2D) which were greatly reduced by Rosi, (2) the stiffness of the 3D organoids which were substantially reduced by Rip or KD025, and (3) TGFß2 induced a significant up-regulation of all ECMs, except for COL6 (2D) or αSMA (3D), and down-regulation of COL6 (2D). Rosi caused a significant up-regulation of COL1, 4 and 6 (3D), and down-regulation of COL6 (2D) and αSMA (3D). Most of these TGFß2-induced expressions in the 2D and αSMA in the 3D were substantially inhibited by KD025, but COL4 and αSMA in 2D were further enhanced by Rip. CONCLUSION: The findings reported herein indicate that TGFß2 induces an increase in fibrogenetic changes on the plane and in the spatial space, and are inhibited by Rosi and ROCK inhibitors, respectively.


Subject(s)
Conjunctiva/metabolism , Fibroblasts/metabolism , Rosiglitazone/pharmacology , Transforming Growth Factor beta2/metabolism , rho-Associated Kinases/antagonists & inhibitors , Actins/biosynthesis , Cell Line , Collagen/biosynthesis , Fibronectins/biosynthesis , Gene Expression Regulation/drug effects , Humans , rho-Associated Kinases/metabolism
4.
Front Endocrinol (Lausanne) ; 12: 645881, 2021.
Article in English | MEDLINE | ID: mdl-34177798

ABSTRACT

Introduction: Assuming myokines underlie some of the health benefits of exercise, we hypothesised that 'high responder trainer' (HRT) rats would exhibit distinct myokine profiles to 'low responder trainers' (LRT), reflecting distinct health and adaptive traits. Methods: Blood was collected from LRT and HRT (N=8) rats at baseline (BL), immediately (0h), 1h, and 3h after running; repeated after 3-wks training. Myokines were analysed by ELISA (i.e. BDNF/Fractalkine/SPARC/Irisin/FGF21/Musclin/IL-6). Results: At baseline, Musclin (LRT: 84 ± 24 vs HRT: 26 ± 3 pg/ml, P=0.05) and FGF21 (LRT: 133 ± 34 vs HRT: 63.5 ± 13 pg/ml, P=0.08) were higher in LRT than HRT. Training increased Musclin in HRT (26 ± 3 to 54 ± 9 pg/ml, P<0.05) and decreased FGF21 in LRT (133 ± 34 to 60 ± 28 pg/ml, P<0.05). Training increased SPARC (LRT: 0.8 ± 0.1 to 2.1 ± 0.6 ng/ml, P<0.05; HRT: 0.7 ± 0.06 to 1.8 ± 0.3 ng/ml, P=0.06) and Irisin (LRT 0.62 ± 0.1 to 2.6 ± 0.4 ng/ml, P<0.01; HRT 0.53 ± 0.1 to 2.8 ± 0.7 ng/ml, P<0.01) while decreasing BDNF (LRT: 2747 ± 293 to 1081 ± 330 pg/ml, P<0.01; HRT: 1976 ± 328 to 797 ± 160 pg/ml, P<0.05). Acute exercise response of Musclin (AUC) was higher in LRT vs HRT (306 ± 74 vs. 88 ± 12 pg/ml×3h-1, P<0.01) and elevated in HRT after training (221 ± 31 pg/ml×3h-1, P<0.01). Training elevated SPARC (LRT: 2.4 ± 0.1 to 7.7 ± 1.3 ng/ml×3h-1, P<0.05; HRT: 2.5 ± 0.13 to 11.2 ± 2.2 ng/ml×3h-1, P<0.001) and Irisin (LRT: 1.34 ± 0.3 to 9.6 ± 1.7 ng/ml×3h-1, P<0.001; HRT: 1.5 ± 0.5 to 12.1 ± 1.9 ng/ml×3h-1, P<0.0001). Conclusion: Exercise training alters how myokines are secreted in response to acute exercise. Myokine responses were not robustly linked to adaptive potential in aerobic capacity, making them an unlikely regulator of adaptive traits.


Subject(s)
Exercise Tolerance , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Running , Animals , Area Under Curve , Brain-Derived Neurotrophic Factor/biosynthesis , Chemokine CX3CL1/biosynthesis , Enzyme-Linked Immunosorbent Assay/methods , Female , Fibroblast Growth Factors/biosynthesis , Fibronectins/biosynthesis , Interleukin-6/biosynthesis , Osteonectin/biosynthesis , Phenotype , Rats , Time Factors , Transcription Factors/biosynthesis
5.
Int J Mol Sci ; 22(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917452

ABSTRACT

Due to the increasing incidence of malignant gliomas, particularly glioblastoma multiforme (GBM), a simple and reliable GBM diagnosis is needed to screen early the death-threaten patients. This study aimed to identify a protein that can be used to discriminate GBM from low-grade astrocytoma and elucidate further that it has a functional role during malignant glioma progressions. To identify proteins that display low or no expression in low-grade astrocytoma but elevated levels in GBM, glycoprotein fibronectin (FN) was particularly examined according to the mining of the Human Protein Atlas. Web-based open megadata minings revealed that FN was mainly mutated in the cBio Cancer Genomic Portal but dominantly overexpressed in the ONCOMINE (a cancer microarray database and integrated data-mining platform) in distinct tumor types. Furthermore, numerous different cancer patients with high FN indeed exhibited a poor prognosis in the PrognoScan mining, indicating that FN involves in tumor malignancy. To investigate further the significance of FN expression in glioma progression, tumor specimens from five malignant gliomas with recurrences that received at least two surgeries were enrolled and examined. The immunohistochemical staining showed that FN expression indeed determined the distinct progressions of malignant gliomas. Furthermore, the expression of vimentin (VIM), a mesenchymal protein that is strongly expressed in malignant cancers, was similar to the FN pattern. Moreover, the level of epithelial-mesenchymal transition (EMT) inducer transforming growth factor-beta (TGF-ß) was almost recapitulated with the FN expression. Together, this study identifies a protein FN that can be used to diagnose GBM from low-grade astrocytoma; moreover, its expression functionally determines the malignant glioma progressions via TGF-ß-induced EMT pathway.


Subject(s)
Brain Neoplasms/metabolism , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Neoplasm Proteins/biosynthesis , Signal Transduction , Transforming Growth Factor beta/metabolism , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Databases, Nucleic Acid , Female , Fibronectins/genetics , Glioblastoma/diagnosis , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Prognosis , Transforming Growth Factor beta/genetics
6.
Front Endocrinol (Lausanne) ; 12: 653179, 2021.
Article in English | MEDLINE | ID: mdl-33897620

ABSTRACT

Obesity is a global health problem and a major risk factor for several metabolic conditions including dyslipidemia, diabetes, insulin resistance and cardiovascular diseases. Obesity develops from chronic imbalance between energy intake and energy expenditure. Stimulation of cellular energy burning process has the potential to dissipate excess calories in the form of heat via the activation of uncoupling protein-1 (UCP1) in white and brown adipose tissues. Recent studies have shown that activation of transforming growth factor-ß (TGF-ß) signaling pathway significantly contributes to the development of obesity, and blockade or inhibition is reported to protect from obesity by promoting white adipose browning and increasing mitochondrial biogenesis. Identification of novel compounds that activate beige/brown adipose characteristics to burn surplus calories and reduce excess storage of fat are actively sought in the fight against obesity. In this review, we present recent developments in our understanding of key modulators of TGF-ß signaling pathways including follistatin (FST) and myostatin (MST) in regulating adipose browning and brown adipose mass and activity. While MST is a key ligand for TGF-ß family, FST can bind and regulate biological activity of several TGF-ß superfamily members including activins, bone morphogenic proteins (BMP) and inhibins. Here, we review the literature supporting the critical roles for FST, MST and other proteins in modulating TGF-ß signaling to influence beige and brown adipose characteristics. We further review the potential therapeutic utility of FST for the treatment of obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Follistatin/biosynthesis , Metabolic Diseases/metabolism , Myostatin/biosynthesis , Obesity/metabolism , Transforming Growth Factor beta1/metabolism , Adipose Tissue, Beige/metabolism , Animals , Energy Metabolism , Fibronectins/biosynthesis , Follistatin/metabolism , Follistatin-Related Proteins/biosynthesis , Humans , Ligands , Mice , Signal Transduction , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
7.
Mol Hum Reprod ; 27(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-33881516

ABSTRACT

Plasma fibronectin 1 (FN1) levels are elevated in individuals with pre-eclampsia (PE), which may be applied as a possible b marker for vascular endothelial injury during PE. In the present study, the possible role of FN1 in the pathogenesis of PE and regulation of apoptosis and autophagy in vascular endothelial cells was explored. Plasma FN1 levels in 80 patients with PE and 40 healthy pregnant individuals were measured using ELISA to verify its relationship with the severity of PE. pcDNA3.1-FN1 or FN1-small interfering (si) RNA was used to manipulate the expression of FN1 in human umbilical vein endothelial cells (HUVECs) to assess the effects of FN1 on cell apoptosis, autophagy, and the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway. It was found that upregulation of FN1 promoted apoptosis and autophagy, in addition to significantly inhibiting the activation of AKT and mTOR in HUVECs. By contrast, downregulation of FN1 expression inhibited cell apoptosis and autophagy, but increased AKT and mTOR phosphorylation in HUVECs that were cultured in serum samples obtained from patients with PE. Rescue experiments found that the PI3K/AKT inhibitor LY294002 reversed the effects of FN1-siRNA on apoptosis and autophagy in HUVECs cultured in serum from patients with PE. Therefore, data from the present study suggest that FN1 participates in the pathogenesis of PE by promoting apoptosis and autophagy in vascular endothelial cells, which is associated with the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Endothelial Cells/pathology , Fibronectins/physiology , Pre-Eclampsia/etiology , Adult , Apoptosis , Autophagy , Case-Control Studies , Chromones/pharmacology , Endothelial Cells/metabolism , Female , Fibronectins/biosynthesis , Fibronectins/blood , Fibronectins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Reporter , Human Umbilical Vein Endothelial Cells , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/pathology , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Severity of Illness Index , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Anticancer Drugs ; 32(7): 693-702, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33675611

ABSTRACT

Lung squamous carcinoma (LUSC) is the second most frequent subtype of non-small cell lung cancer. Rarely gene alterations are identified in LUSC. Therefore, identifying LUSC-related genes to explain the relevant molecular mechanism is urgently needed. A potential biomarker, calcium-activated nucleotidase 1 (CANT1), was elevated in tissues of LUSC patients relative to normal cases based on the TCGA and/or GTEx database. CCK-8 and transwell tests were then implemented to measure the proliferative, invasive and migratory capacities, and showed that knockdown of CANT1 blocked LUSC cells proliferation. miR-607, predicted as an upstream factor for CANT1, was declined in LUSC using TargetScan analysis and luciferase activity test. Low miR-607 expression was related with unfavorable outcomes of LUSC patients. Moreover, miR-607 downregulation elevated cell viability, invasion and migration in LUSC cells, which was antagonized by si-CANT1. GEPIA website was accessed to estimate the relevance between CANT1 and epithelial-mesenchymal transition (EMT)-related positive factors. The protein levels of Fibronectin, Vimentin, Snail and ß-catenin were altered due to the abnormal CANT1 and miR-607 expression. Together, these data unveiled that miR-607/CANT1 pair may exert a vital role in the progression of LUSC through mediating EMT process, which would furnish an available therapeutic therapy for LUSC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , MicroRNAs/metabolism , Nucleotidases/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Survival , Down-Regulation , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Vimentin/biosynthesis
9.
Int J Biol Macromol ; 178: 316-324, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33652046

ABSTRACT

Irisin is a muscle factor discovered in 2012 that plays an important role in many tissues, including bone. Eight years since its discovery, there are still many controversies regarding its molecular biology, detection, and effects on bone. This article summarizes the points raised to date, and discusses the mechanisms by which irisin regulates bone cells. The information reviewed here provides a useful foundation for future research.


Subject(s)
Bone and Bones/metabolism , Fibronectins/biosynthesis , Osteocytes/metabolism , Humans
10.
Cancer Lett ; 508: 59-72, 2021 06 28.
Article in English | MEDLINE | ID: mdl-33771684

ABSTRACT

The loss of cell-matrix interactions induces apoptosis, known as anoikis. For successful distant metastasis, circulating tumor cells (CTCs) that have lost matrix attachment need to acquire anoikis resistance in order to survive. Cell aggregate formation confers anoikis resistance, and CTC clusters are more highly metastatic compared to single cells; however, the molecular mechanisms underlying this aggregation are not well understood. In this study, we demonstrated that cell detachment increased cell aggregation and upregulated fibronectin (FN) levels in lung and breast cancer cells, but not in their normal counterparts. FN knockdown decreased cell aggregation and increased anoikis. In addition, cell detachment induced cell-cell adhesion proteins, including E-cadherin, desmoglein-2, desmocollin-2/3, and plakoglobin. Interestingly, FN knockdown decreased the levels of desmoglein-2, desmocollin-2/3, and plakoglobin, but not E-cadherin, suggesting the involvement of desmosomal junction in cell aggregation. Accordingly, knockdown of desmoglein-2, desmocollin-2, or plakoglobin reduced cell aggregation and increased cell sensitivity to anoikis. Previously, we reported that NADPH oxidase 4 (Nox4) upregulation is important for anoikis resistance. Nox4 inhibition by siRNA or apocynin decreased cell aggregation and increased anoikis with the downregulation of FN, and, consequently, decreased desmoglein-2, desmocollin-2/3, or plakoglobin. The coexpression of Nox4 and FN was found to be significant in lung and breast cancer patients, based on cBioPortal data. In vivo mouse lung metastasis model showed that FN knockdown suppressed lung metastasis and thus enhanced survival. FN staining of micro tissue array revealed that FN expression was positive for human lung cancer (61%) and breast cancer (58%) patients. Furthermore, the expression levels of FN, desmoglein-2, desmocollin-2, and plakoglobin were significantly correlated with the poor survival of lung and breast cancer patients, as per the Kaplan-Meier plotter analysis. Altogether, our data suggest that FN upregulation and enhanced desmosomal interactions are critical for cell aggregation and anoikis resistance upon cell detachment.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibronectins/biosynthesis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , A549 Cells , Animals , Anoikis/physiology , Breast Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Aggregation/physiology , Cell Line, Tumor , Fibronectins/genetics , Fibronectins/metabolism , Heterografts , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Mice , Mice, Nude , NADPH Oxidase 4/biosynthesis , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Neoplasm Metastasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Array Analysis , Up-Regulation
11.
Biomolecules ; 11(2)2021 02 20.
Article in English | MEDLINE | ID: mdl-33672565

ABSTRACT

Irisin is an exercise-induced myokine, suggested to exert beneficial effects on metabolism. However, the studies on the regulation of irisin secretion and the expression of its precursor FNDC5 have shown conflicting data. The discrepancies among previous correlation studies in humans are related to the heterogeneity of the study population. The fact that irisin is not only a myokine but also an adipokine leads to the further complexity of the role of irisin in metabolic regulation. In this study, we examined the regulation of FNDC5 expression and irisin in circulation in both type 1 and type 2 diabetic mice, and their potential relationships with metabolic parameters. In streptozotocin (STZ)-induced type 1 diabetic mice, high-fat diet (HFD)-induced obese mice and db/db mice, the circulating irisin as well as FNDC5 gene expression in subcutaneous fat was downregulated. Muscle FNDC5 expression was only significantly lower in STZ mice, and epididymal fat FNDC5 expression was unaltered. It is interesting to note that plasma irisin levels correlated positively with subcutaneous fat FNDC5 expression, but not epididymal fat or muscle. Moreover, both irisin levels and subcutaneous fat FNDC5 correlated negatively with markers of insulin resistance. These results suggest a regulatory role for subcutaneous fat-derived FNDC5/irisin in metabolic disease.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Fibronectins/biosynthesis , Fibronectins/blood , Metabolic Syndrome/metabolism , Muscle, Skeletal/metabolism , Adipokines/metabolism , Animals , Blood Glucose/metabolism , Disease Models, Animal , Gene Expression Regulation , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
12.
Front Immunol ; 12: 754475, 2021.
Article in English | MEDLINE | ID: mdl-35003066

ABSTRACT

Nonresolving inflammation is a critical driver of several chronic inflammatory diseases, including inflammatory bowel diseases (IBD). This unresolved inflammation may result from the persistence of an initiating stimulus or from the alteration of the resolution phase of inflammation. Elimination of apoptotic cells by macrophages (a process called efferocytosis) is a critical step in the resolution phase of inflammation. Efferocytosis participates in macrophage reprogramming and favors the release of numerous pro-resolving factors. These pro-resolving factors exert therapeutic effects in experimental autoimmune arthritis. Here, we propose to evaluate the efficacy of pro-resolving factors produced by macrophages after efferocytosis, a secretome called SuperMApo, in two IBD models, namely dextran sodium sulfate (DSS)-induced and T cell transfer-induced colitis. Reintroducing these pro-resolving factors was sufficient to decrease clinical, endoscopic and histological colitis scores in ongoing naive T cell-transfer-induced colitis and in DSS-induced colitis. Mouse primary fibroblasts isolated from the colon demonstrated enhanced healing properties in the presence of SuperMApo, as attested by their increased migratory, proliferative and contractive properties. This was confirmed by the use of human fibroblasts isolated from patients with IBD. Exposure of an intestinal epithelial cell (IEC) line to these pro-resolving factors increased their proliferative properties and IEC acquired the capacity to capture apoptotic cells. The improvement of wound healing properties induced by SuperMApo was confirmed in vivo in a biopsy forceps-wound colonic mucosa model. Further in vivo analysis in naive T cell transfer-induced colitis model demonstrated an improvement of intestinal barrier permeability after administration of SuperMApo, an intestinal cell proliferation and an increase of α-SMA expression by fibroblasts, as well as a reduction of the transcript coding for fibronectin (Fn1). Finally, we identified TGF-ß, IGF-I and VEGF among SuperMApo as necessary to favor mucosal healing and confirmed their role both in vitro (using neutralizing antibodies) and in vivo by depleting these factors from efferocytic macrophage secretome using antibody-coated microbeads. These growth factors only explained some of the beneficial effects induced by factors released by efferocytic macrophages. Overall, the administration of pro-resolving factors released by efferocytic macrophages limits intestinal inflammation and enhance tissue repair, which represents an innovative treatment of IBD.


Subject(s)
Biological Factors/physiology , Cytophagocytosis/physiology , Fibroblasts/physiology , Inflammatory Bowel Diseases/immunology , Macrophages/physiology , Wound Healing/physiology , Actins/biosynthesis , Actins/genetics , Animals , Biological Factors/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Division/drug effects , Cell Line , Colitis/chemically induced , Colitis/etiology , Colitis/immunology , DNA-Binding Proteins/deficiency , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Fibronectins/biosynthesis , Fibronectins/genetics , Humans , Inflammatory Bowel Diseases/physiopathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/cytology , Intestinal Mucosa/injuries , Lymphocyte Transfusion/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms
13.
Rom J Morphol Embryol ; 62(3): 705-712, 2021.
Article in English | MEDLINE | ID: mdl-35263398

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an essential biological process involved in the initiation and progression of cancer by which epithelial tumor cells lose their differentiated characteristics, such as cell-cell adhesion and apical-basal polarity and acquire a more invasive and∕or metastatic mesenchymal phenotype. The present study investigated the expression of immunomarkers with a role in EMT of non-melanoma skin cancers (NMSCs), such as E-cadherin, fibronectin and Slug, for a number of 50 NMSCs, represented by 30 cases of basal cell carcinomas (BCCs) and 20 cases of squamous cell carcinomas (SCCs). For BCC, the statistical analysis of the investigated immunomarkers indicated significantly differences in relation to the depth of invasion, and for E-cadherin and fibronectin with the degree of risk. In the case of SCC, the statistical analysis indicated significant differences of E-cadherin and Slug with the degree of tumor differentiation, and for fibronectin and Slug with the depth of invasion. The analysis of the distribution for the percentage values of the investigated immunomarkers in the case of BCC indicated a significant negative linear relation between E-cadherin/fibronectin and E-cadherin/Slug, and in SCC a significant negative linear relation between E-cadherin∕fibronectin, E-cadherin∕Slug and a positive linear one in the case of fibronectin∕Slug. The study indicates through the statistically significant relation between E-cadherin∕fibronectin and E-cadherin∕Slug, the EMT intervention in carcinogenesis of NMSC.


Subject(s)
Antigens, CD , Cadherins , Fibronectins , Skin Neoplasms , Snail Family Transcription Factors , Antigens, CD/biosynthesis , Antigens, CD/immunology , Cadherins/biosynthesis , Cadherins/immunology , Epithelial-Mesenchymal Transition , Fibronectins/biosynthesis , Fibronectins/immunology , Gene Expression Regulation, Neoplastic , Humans , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Snail Family Transcription Factors/biosynthesis , Snail Family Transcription Factors/immunology
14.
Matrix Biol ; 94: 31-56, 2020 12.
Article in English | MEDLINE | ID: mdl-32777343

ABSTRACT

Although the matricellular protein periostin is prominently upregulated in skin and gingival healing, it plays contrasting roles in myofibroblast differentiation and matrix synthesis respectively. Palatal healing is associated with scarring that can alter or restrict maxilla growth, but the expression pattern and contribution of periostin in palatal healing is unknown. Using periostin-knockout (Postn-/-) and wild-type (WT) mice, the contribution of periostin to palatal healing was investigated through 1.5 mm full-thickness excisional wounds in the hard palate. In WT mice, periostin was upregulated 6 days post-wounding, with mRNA levels peaking at day 12. Genetic deletion of periostin significantly reduced wound closure rates compared to WT mice. Absence of periostin reduced mRNA levels of pivotal genes in wound repair, including α-SMA/acta2, fibronectin and ßigh3. Recruitment of fibroblasts and inflammatory cells, as visualized by immunofluorescent staining for fibroblast specific factor-1, vimentin, and macrophages markers Arginase-1 and iNOS was also impaired in Postn-/-, but not WT mice. Palatal fibroblasts isolated from the hard palate of mice were cultured on collagen gels and prefabricated silicon substrates with varying stiffness. Postn-/- fibroblasts showed a significantly reduced ability to contract a collagen gel, which was rescued by the exogenous addition of recombinant periostin. As the stiffness increased, Postn-/- fibroblasts increasingly differentiated into myofibroblasts, but not to the same degree as the WT. Pharmacological inhibition of Rac rescued the deficient myofibroblastic phenotype of Postn-/- cells. Low stiffness substrates (0.2 kPa) resulted in upregulation of fibronectin in WT cells, an effect which was significantly reduced in Postn-/- cells. Quantification of immunostaining for vinculin and integrinß1 adhesions revealed that Periostin is required for the formation of focal and fibrillar adhesions in mPFBs. Our results suggest that periostin modulates myofibroblast differentiation and contraction via integrinß1/RhoA pathway, and fibronectin synthesis in an ECM stiffness dependent manner in palatal healing.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Fibronectins/genetics , Palate, Hard/growth & development , Wound Healing/genetics , Actins/genetics , Animals , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/biosynthesis , Humans , Integrin beta1/genetics , Maxilla/growth & development , Maxilla/metabolism , Mice , Mice, Knockout , Myofibroblasts/metabolism , Myofibroblasts/pathology , Palate, Hard/metabolism , Palate, Hard/physiopathology , Signal Transduction/genetics , rhoA GTP-Binding Protein/genetics
15.
Knee ; 27(3): 755-759, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32563433

ABSTRACT

BACKGROUND: Enhanced chondrogenesis and reduction in hypertrophy are essential pre-requisites for cell-based therapy in regenerative research for cartilage loss. Chondroprogenitors, isolated by fibronectin adhesion assay (FAA), have shown promising results in various preclinical studies due to their inherent characteristics. However, the need for monolayer culture and the effect of expansion on cell phenotype render differentiation between chondroprogenitors and chondrocytes (native cartilage cells) difficult. This is further complicated due to reported de-differentiation of chondrocytes in culture. Thus, the aim of our study was to harvest cells from articular cartilage and compare their gene expression to cells demonstrating adherence and non-adherence to fibronectin. METHOD: Fresh-cells (FC) were isolated from human osteoarthritic knee joints(n = 3) and subjected to FAA. Cells unbound to fibronectin (20 min after plating) were termed as FAA-ve. Attached cells were further cultured for five population doublings and designated FAA+ve. RNA from all three cell groups was assessed for SOX-9, ACAN, COL2A1, COL1A1, RUNX2 and COL10A1. RESULTS: All three groups exhibited moderate to high expression of markers of chondrogenesis and marker of chondrocyte hypertrophy. FAA+ve group exhibited significantly lower levels of hypertrophy markers: RUNX2 (vs FC and FAA-ve, P = 0.018) and COL10A1(vs FAA-ve, P = 0.005). CONCLUSIONS: Our results demonstrated that fibronectin effectively isolated cells distinct from mature chondrocytes in terms of reduced hypertrophic tendency. This is noteworthy as cells isolated by FAA, retaining their inherent progenitor phenotype, with upregulation of chondrogenic markers may be used successfully for cartilage repair in future translational work.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/cytology , Chondrogenesis/genetics , DNA/genetics , Fibronectins/genetics , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/metabolism , Female , Fibronectins/biosynthesis , Humans , Male , Middle Aged
16.
Biomed Pharmacother ; 127: 110119, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32276127

ABSTRACT

Idiopathic pulmonary fibrosis (IPF), characterized by excessive collagen deposition, is a progressive and typically fatal lung disease without effective therapeutic strategies. Juglanin, as a natural product mainly isolated from green walnut husks of Juglans mandshuric, has various bioactivities, including anti-oxidative, anti-inflammatory and anti-fibrotic effects. Stimulator of interferon genes (Sting) is a signaling molecule and plays an essential role in meditating fibrosis. However, the effects of Jug and Sting on pulmonary fibrosis are not fully understood. In this study, we investigated the role of Jug in bleomycin (BLM)-induced inflammation and fibrosis mouse model, as well as the underlying molecular mechanism. The results here indicated that Jug-treated mice exhibited a definitively improved survival rate than that of the BLM-challenged mice. Jug administration significantly alleviated neutrophil alveolar infiltration, lung vascular permeability and pro-inflammatory response in BLM mice. Subsequently, the pulmonary fibrosis induced by BLM was markedly attenuated by Jug through reducing the expression of fibrotic hallmarks, including transforming growth factor-ß1 (TGF-ß1), fibronectin, matrix metallo-proteinase-9 (MMP-9), α-smooth muscle actin (α-SMA) and collagen I. Importantly, we found that BLM mice showed higher expression levels of Sting in lung tissues, which were notably restrained by Jug treatment. The role of Jug in suppressing Sting was confirmed in TGF-ß-incubated cells. Notably, the in vitro analysis further showed that Sting knockdown could ameliorate TGF-ß-triggered collagen accumulation. In contrast, TGF-ß-induced fibrosis was accelerated by Sting over-expression. Therefore, BLM may induce lung fibrosis through activating Sting signaling, and Jug could be used therapeutically to improve tissue repair and attenuate the intractable disease.


Subject(s)
Glycosides/pharmacology , Inflammation/prevention & control , Kaempferols/pharmacology , Membrane Proteins/metabolism , Pulmonary Fibrosis/prevention & control , Actins/biosynthesis , Animals , Bleomycin , Collagen Type I/biosynthesis , Fibronectins/biosynthesis , Gene Knockdown Techniques , Inflammation/complications , Male , Matrix Metalloproteinase 9/biosynthesis , Membrane Proteins/genetics , Mice , Pulmonary Fibrosis/chemically induced , Signal Transduction/drug effects , Survival Rate , Transforming Growth Factor beta1/biosynthesis
17.
Technol Cancer Res Treat ; 19: 1533033820909911, 2020.
Article in English | MEDLINE | ID: mdl-32281480

ABSTRACT

OBJECTIVE: Nasopharyngeal carcinoma is highly endemic in Southeast China. Circulating tumor cell is an important biomarker in the prognosis of variety kinds of cancers. Overexpression of fibronectin 1 was observed in variety kinds of malignancies and may contribute to progress and metastasis of the cancers. The current study was aimed to investigate phenotypes of circulating tumor cell in nasopharyngeal carcinoma blood and fibronectin 1 expression in the circulating tumor cell, and their clinical application in predicting nasopharyngeal carcinoma prognosis. METHODS: Blood samples were obtained from nasopharyngeal carcinoma patients before and after treatment. CanPatrol circulating tumor cell enrichment and RNA in situ hybridization were applied to identify circulating tumor cell and its phenotypes. Fibronectin 1 messenger RNA expression in the cells of circulating tumors was examined by messenger RNA-in situ hybridization. RESULTS: Circulating tumor cell was not associated with tumor characteristics or lymph node metastasis. Patients with >9 circulating tumor cells or >5 mesenchymal phenotype circulating tumor cell per 5-mL blood had poorer progression-free survival (P < .05). Multivariable analysis demonstrated that 2 or more mesenchymal phenotype circulating tumor cells with high fibronectin 1 messenger RNA expression predicted shorter progression-free survival (P < .05). CONCLUSIONS: Circulating tumor cells with high-level fibronectin 1 expression was associated with poor survival in patients with nasopharyngeal carcinoma and could be an independent prognostic factor for nasopharyngeal carcinoma.


Subject(s)
Fibronectins/biosynthesis , Nasopharyngeal Carcinoma/blood , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/blood , Nasopharyngeal Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Aged, 80 and over , Female , Fibronectins/genetics , Fibronectins/metabolism , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Neoplastic Cells, Circulating/pathology , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
18.
Eur J Dermatol ; 30(1): 3-11, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32250253

ABSTRACT

BACKGROUND: Platelet lysate (PL) contains a cocktail of growth factors and cytokines that promote tissue repair and regeneration. In vitro studies have shown that PL may affect the reparative function of keratinocytes and fibroblasts, but little is known about the effect of PL on immune cells involved in wound healing. OBJECTIVES: To analyse the effects of PL on T cells involved in the wound repair process. MATERIALS AND METHODS: The effect of PL on T cell proliferation, activation, and cytokine production was measured by ELISA and cytofluorometry and regulatory function based on cytofluorometry and Foxp3 RNA expression. Using an in vitro model of wound healing, we investigated the effect of PL-treated T cells on fibroblast proliferation and production of fibronectin and type-1 collagen as well as keratinocyte migration. RESULTS: PL induced T lymphocyte proliferation and CD69 expression, and promoted a transient upregulation of IFN-γ and TNF-α. However, later on, PL enhanced the number of CD25+ T cells releasing TGF-ß and expressing Foxp3 RNA, which was accompanied by a suppression in the level of type 1 cytokines. In the in vitro model, supernatants of PL-treated T cells positively affected the reparative capacity of human keratinocytes and induced fibroblast proliferation and production of fibronectin and type-1 collagen. CONCLUSION: These results indicate that PL temporally regulates T cells during the healing process, enhancing protective cytokines in the early phase, followed by a prominent expansion of TGF-ß+ T regulatory cells that promote tissue regeneration and dampen the inflammatory response to prevent excessive tissue damage.


Subject(s)
Blood Platelets , Fibroblasts/metabolism , Keratinocytes/physiology , RNA/metabolism , T-Lymphocytes, Regulatory/metabolism , Wound Healing , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/biosynthesis , Fibronectins/biosynthesis , Forkhead Transcription Factors/genetics , Humans , In Vitro Techniques , Interferon-gamma/biosynthesis , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/physiology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
19.
Neurosci Lett ; 725: 134902, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32165261

ABSTRACT

Oxidative stress is implicated in the initiation and progression of human and animal diseases. MicroRNA (MiR) has been reported to be involved in the body's regulation to oxidative stress. We investigated if miR-183 regulates lipopolysaccharide (LPS)-induced oxidative stress in the hippocampus of weaned piglets. LPS-treated piglets had lower expression of miR-183 and higher expression of the fibronectin(FN)1 gene in their hippocampus than control piglets. The expression profiles of miR-183 and the FN1 gene in primary cultured rat hippocampal neurons exposed to LPS were consistent with those in the hippocampus of LPS-treated piglets. The LPS-induced expression of FN1 was reversed in hippocampal neurons by transfection with an miR-183 mimic. A luciferase reporter assay further demonstrated that the FN1 gene is a direct target of miR-183. Taken together, our results demonstrated that miR-183 regulates LPS-induced oxidative stress at least in part by targeting FN1.


Subject(s)
Fibronectins/biosynthesis , Hippocampus/metabolism , Lipopolysaccharides/toxicity , MicroRNAs/biosynthesis , Neurons/metabolism , Oxidative Stress/physiology , Animals , Animals, Newborn , Hippocampus/drug effects , Male , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Swine
20.
Medicine (Baltimore) ; 99(8): e19235, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32080125

ABSTRACT

Type 2 diabetes is the fastest growing metabolic disease in the world. Recently, muscle is considered an endocrine organ which secretes various peptides that play an important role in insulin resistance and metabolic syndrome. We assessed 4 different myokines, irisin, interleukin-13 (IL-13), follistatin-related protein-1 (FSTL-1), and fractalkine, in normal, prediabetes, and diabetes patients.A total of 126 participants who visited Gangnam Severance Hospital were enrolled and divided into normal, prediabetes, and diabetes groups based on oral glucose tolerance test and hemoglobin a1c. A cross-sectional study was conducted to measure and compare serum levels of irisin, IL-13, FSTL-1, and fractalkine among the groups.Irisin level showed a tendency to increase in prediabetes group compared to normal group (P < .1) but showed a significant decrease when comparing diabetes from prediabetes group (P < .001). IL-13 decreased in diabetes group compared to prediabetes and normal group (P < .001, P < .05, respectively). FSTL-1 of diabetes group was lower than that of prediabetes group (P < .05), and fractalkine was higher in diabetes group compared to that of prediabetes and normal group (P < .01, P < .01, respectively).Irisin, IL-13, and FSTL-1 levels were reduced in diabetes group compared to normal or prediabetes group while fractalkine showed a progressive increase from normal to diabetes group. Further studies are warranted to study the roles of various myokine in diabetes through a larger prospective study.


Subject(s)
Cytokines/biosynthesis , Diabetes Mellitus, Type 2/physiopathology , Fibronectins/biosynthesis , Follistatin-Related Proteins/biosynthesis , Prediabetic State/physiopathology , Adult , Aged , Chemokine CX3CL1/biosynthesis , Cross-Sectional Studies , Female , Glucose Intolerance/physiopathology , Glycated Hemoglobin , Humans , Male , Middle Aged , Prospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...