Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.380
Filter
1.
Cardiovasc Diabetol ; 23(1): 298, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143620

ABSTRACT

BACKGROUND: Activation of brown adipose tissue (BAT) has gained attention due to its ability to dissipate energy and counteract cardiometabolic diseases (CMDs). METHODS: This study investigated the consequences of cold exposure on the BAT and liver proteomes of an established CMD mouse model based on LDL receptor-deficient (LdlrKO) mice fed a high-fat, high-sucrose, high-cholesterol diet for 16 weeks. We analyzed energy metabolism in vivo and performed untargeted proteomics on BAT and liver of LdlrKO mice maintained at 22 °C or 5 °C for 7 days. RESULTS: We identified several dysregulated pathways, miRNAs, and transcription factors in BAT and liver of cold-exposed Ldlrko mice that have not been previously described in this context. Networks of regulatory interactions based on shared downstream targets and analysis of ligand-receptor pairs identified fibrinogen alpha chain (FGA) and fibronectin 1 (FN1) as potential crosstalk factors between BAT and liver in response to cold exposure. Importantly, genetic variations in the genes encoding FGA and FN1 have been associated with cardiometabolic-related phenotypes and traits in humans. DISCUSSION: This study describes the key factors, pathways, and regulatory networks involved in the crosstalk between BAT and the liver in a cold-exposed CMD mouse model. These findings may provide a basis for future studies aimed at testing whether molecular mediators, as well as regulatory and signaling mechanisms involved in tissue adaption upon cold exposure, could represent a target in cardiometabolic disorders.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Disease Models, Animal , Energy Metabolism , Gene Regulatory Networks , Liver , Mice, Knockout , Proteomics , Receptors, LDL , Signal Transduction , Animals , Adipose Tissue, Brown/metabolism , Liver/metabolism , Energy Metabolism/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, LDL/deficiency , Male , Fibrinogen/metabolism , Fibrinogen/genetics , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Fibronectins/metabolism , Fibronectins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Mice , Gene Expression Regulation , Protein Interaction Maps
2.
Physiol Rep ; 12(15): e16179, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107084

ABSTRACT

Diabetic kidney disease (DKD) is a devastating kidney disease and lacks effective therapeutic interventions. The present study was aimed to determine whether reconstituted high-density lipoprotein (rHDL) ameliorated renal injury in eNOS-/- dbdb mice, a mouse model of DKD. Three groups of mice, wild type C57BLKS/J (non-diabetes), eNOS-/- dbdb (diabetes), and eNOS-/- dbdb treated with rHDL (diabetes+rHDL) with both males and females were used. The rHDL nanoparticles were administered to eNOS-/- dbdb mice at Week 16 at 5 µg/g body weight in ~100 µL of saline solution twice per week for 4 weeks via retroorbital injection. We found that rHDL treatment significantly blunted progression of albuminuria and GFR decline observed in DKD mice. Histological examinations showed that the rHDLs significantly alleviated glomerular injury and renal fibrosis, and inhibited podocyte loss. Western blots and immunohistochemical examinations showed that increased protein abundances of fibronectin and collagen IV in the renal cortex of eNOS-/- dbdb mice were significantly reduced by the rHDLs. Taken together, the present study suggests a renoprotective effect of rHDLs on DKD.


Subject(s)
Diabetic Nephropathies , Lipoproteins, HDL , Mice, Inbred C57BL , Nitric Oxide Synthase Type III , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Mice , Male , Nitric Oxide Synthase Type III/metabolism , Lipoproteins, HDL/pharmacology , Female , Mice, Knockout , Kidney/pathology , Kidney/metabolism , Kidney/drug effects , Albuminuria , Fibronectins/metabolism , Fibronectins/genetics , Fibrosis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy
3.
J Immunother Cancer ; 12(8)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39142716

ABSTRACT

BACKGROUND: Anti-PD-1 antibodies have revolutionized cancer immunotherapy due to their ability to induce long-lasting complete remissions in a proportion of patients. Current research efforts are attempting to identify biomarkers and suitable combination partners to predict or further improve the activity of immune checkpoint inhibitors. Antibody-cytokine fusions are a class of pharmaceuticals that showed the potential to boost the anticancer properties of other immunotherapies. Extradomain A-fibronectin (EDA-FN), which is expressed in most solid and hematological tumors but is virtually undetectable in healthy adult tissues, is an attractive target for the delivery of cytokine at the site of the disease. METHODS: In this work, we describe the generation and characterization of a novel interleukin-7-based fusion protein targeting EDA-FN termed F8(scDb)-IL7. The product consists of the F8 antibody specific to the alternatively spliced EDA of FN in the single-chain diabody (scDb) format fused to human IL-7. RESULTS: F8(scDb)-IL7 efficiently stimulates human peripheral blood mononuclear cells in vitro. Moreover, the product significantly increases the expression of T Cell Factor 1 (TCF-1) on CD8+T cells compared with an IL2-fusion protein. TCF-1 has emerged as a pivotal transcription factor that influences the durability and potency of immune responses against tumors. In preclinical cancer models, F8(scDb)-IL7 demonstrates potent single-agent activity and eradicates sarcoma lesions when combined with anti-PD-1. CONCLUSIONS: Our results provide the rationale to explore the combination of F8(scDb)-IL7 with anti-PD-1 antibodies for the treatment of patients with cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Fibronectins , Interleukin-7 , Humans , Fibronectins/metabolism , Fibronectins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Interleukin-7/metabolism , Interleukin-7/pharmacology , Animals , Mice , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Up-Regulation , Female , Cell Line, Tumor
4.
Matrix Biol ; 132: 47-58, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147560

ABSTRACT

BACKGROUND: Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-ß (TGF-ß) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS: Stimulation of lung fibroblasts with TGF-ß1 and TGF-ß2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-ß1 and TGF-ß2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-ß1 or TGF-ß2. Mechanistically, IL-1α administration led to the suppression of TGF-ß1 and TGF-ß2 signaling, through downregulation of mRNA and protein for TGF-ß receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION: IL-1α acts as a master regulator, modulating TGF-ß1 and TGF-ß2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-ß signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS: Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-ß1 or TGF-ß2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.


Subject(s)
Extracellular Matrix , Fibroblasts , Interleukin-1alpha , Lung , Signal Transduction , Transforming Growth Factor beta1 , Humans , Interleukin-1alpha/metabolism , Interleukin-1alpha/genetics , Extracellular Matrix/metabolism , Transforming Growth Factor beta1/metabolism , Lung/metabolism , Lung/cytology , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/cytology , Cell Differentiation , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Cells, Cultured , Collagen Type I/metabolism , Collagen Type I/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Fibronectins/metabolism , Fibronectins/genetics , Gene Expression Regulation/drug effects , Transforming Growth Factor beta2
5.
Commun Biol ; 7(1): 907, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068227

ABSTRACT

Affilin proteins, artificial binding proteins based on the ubiquitin scaffold, have been generated by directed protein evolution to yield de-novo variants that bind the extra-domain B (EDB) of oncofetal fibronectin, an established marker of tumor neovasculature. The crystal structures of two EDB-specific Affilin variants reveal a striking structural plasticity of the ubiquitin scaffold, characterised by ß-strand slippage, leading to different negative register shifts of the ß5 strands. This process recruits amino acid residues from ß5 towards the N-terminus to an adjacent loop region and subsequent residues into ß5, respectively, remodeling the binding interface and leading to target specificity and affinity. Protein backbone alterations resulting from ß-strand register shifts, as seen in the ubiquitin fold, can pose additional challenges to protein engineering as structural evidence of these events is still limited and they are difficult to predict. However, they can surface under the selection pressure of directed evolution and suggest that backbone plasticity allowing ß-strand slippages can increase structural diversity, enhancing the evolutionary potential of a protein scaffold.


Subject(s)
Fibronectins , Ubiquitin , Fibronectins/metabolism , Fibronectins/chemistry , Fibronectins/genetics , Ubiquitin/metabolism , Humans , Protein Binding , Protein Conformation, beta-Strand , Models, Molecular , Crystallography, X-Ray , Protein Engineering
6.
Shanghai Kou Qiang Yi Xue ; 33(2): 123-129, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-39005086

ABSTRACT

PURPOSE: To investigate the effect of Morinda officinalis polysaccharides(MOP) on the expression of fibronectin(FN) and fibronectin containing extra domain A(FN-EDA) in inflammatory periodontal ligament fibroblasts. METHODS: Thirty six rats were randomly divided into a control group(n=12) and a model group (n=24). The model group used orthodontic wire ligation to establish periodontitis. After three weeks, 6 rats from each group were selected and confirmed by Micro-CT to complete the modeling. The remaining rats in the model group were randomly divided into periodontitis group, normal saline(NS) group, and MOP group. In the MOP group, MOP (200 mg/kg for 3 d, 50 µL for 4 weeks) was injected into the palatal side of the left maxillary first molar of the rats. In the NS group, same volume of NS was injected, and no treatment was performed in the periodontitis group. The left maxillary tissue of rats were taken and the pathological changes of periodontal tissue were observed by H-E staining. The expression of FN and FN-EDA was detected by immunohistochemistry. Periodontal ligament fibroblasts were cultured in vitro, the effect of MOP on cell activity detected by CCK-8. The fourth generation cells were divided into control group, inflammation group (10 mg/mL lipopolysaccharide), and experimental group (12.5 µg/mL MOP, 12.5 µg/mL MOP+10 mg/mL lipopolysaccharide). The expression of FN and FN-EDA was detected by qRT-PCR and Western blot. The data were statistically analyzed using Prism 8.0 software package. RESULTS: In vivo experiments, the expression of FN-EDA in the MOP group was significantly lower than that in the periodontitis group and NS group(P<0.05), and the infiltration of inflammatory cells was reduced. However, there was no significant difference in the expression of FN in each group. In vitro experiments, compared with the control group, the expression of FN-EDA mRNA and protein in the inflammation group was significantly increased(P<0.000 1). MOP significantly reduced the expression of FN-EDA in inflammatory cells, but had no significant effect on FN expression. CONCLUSIONS: With increased expression of FN-EDA in inflammatory periodontal ligament tissues and cells, MOP may play a role in inhibiting inflammation by down-regulating FN-EDA.


Subject(s)
Fibroblasts , Fibronectins , Morinda , Periodontal Ligament , Polysaccharides , Animals , Fibroblasts/drug effects , Fibroblasts/metabolism , Polysaccharides/pharmacology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Periodontal Ligament/cytology , Rats , Morinda/chemistry , Fibronectins/metabolism , Fibronectins/genetics , Periodontitis/drug therapy , Periodontitis/metabolism , Inflammation/drug therapy , Rats, Sprague-Dawley
7.
Am J Surg Pathol ; 48(8): 991-1004, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39016330

ABSTRACT

Recently, FN1 fusions to receptor tyrosine kinase genes have been identified in soft tissue tumors with calcified chondroid matrix named calcifying chondroid mesenchymal neoplasms (CCMNs). We collected 33 cases of CCMN from the French network for soft tissue and bone tumors. We performed whole-exome RNA sequencing, expression analysis, and genome-wide DNA methylation profiling in 33, 30, and 20 cases of CCMN compared with a control group of tumors, including noncalcified tenosynovial giant cell tumor (TGCT). Among them, 15 cases showed morphologic overlap with soft tissue chondroma, 8 cases with tophaceous pseudogout, and 10 cases with chondroid TGCT. RNA-sequencing revealed a fusion of FN1 in 76% of cases (25/33) with different 5' partners, including most frequently FGFR2 (14 cases), TEK or FGFR1. Among CCMN associated with FGFR1 fusions, 2 cases had overexpression of FGF23 without tumor-induced osteomalacia. Four CCMN had PDGFRA::USP8 fusions; 3 of which had histologic features of TGCT and were located in the hip, foot, and temporomandibular joint (TMJ). All cases with FN1::TEK fusion were located at TMJ and had histologic features of TGCT with or without chondroid matrix. They formed a distinct cluster on unsupervised clustering analyses based on whole transcriptome and genome-wide methylome data. Our study confirms the high prevalence of FN1 fusions in CCMN. In addition, through transcriptome and methylome analyses, we have identified a novel subgroup of tumors located at the TMJ, exhibiting TGCT-like features and FN1::TEK fusions.


Subject(s)
Biomarkers, Tumor , Calcinosis , Fibroblast Growth Factor-23 , Humans , Female , Male , Middle Aged , Calcinosis/genetics , Calcinosis/pathology , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Young Adult , DNA Methylation , Adolescent , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Fibronectins/genetics , Exome Sequencing , Child , Aged, 80 and over , France , Phenotype
8.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000197

ABSTRACT

Molecular genetic events are among the numerous factors affecting the clinical course of papillary thyroid carcinoma (PTC). Recent studies have demonstrated that aberrant expression of miRNA, as well as different thyroid-related genes, correlate with the aggressive clinical course of PTC and unfavorable treatment outcomes, which opens up new avenues for using them in the personalization of the treatment strategy for patients with PTC. In the present work, our goal was to assess the applicability of molecular markers in the preoperative diagnosis of aggressive variants of papillary thyroid cancer. The molecular genetic profile (expression levels of 34 different markers and BRAF mutations) was studied for 108 cytology specimens collected by fine-needle aspiration biopsy in patients with PTC having different clinical manifestations. Statistically significant differences with adjustment for multiple comparisons (p < 0.0015) for clinically aggressive variants of PTC were obtained for four markers: miRNA-146b, miRNA-221, fibronectin 1 (FN1), and cyclin-dependent kinase inhibitor 2A (CDKN2A) genes. A weak statistical correlation (0.0015 < p < 0.05) was observed for miRNA-31, -375, -551b, -148b, -125b, mtDNA, CITED1, TPO, HMGA2, CLU, NIS, SERPINA1, TFF3, and TMPRSS4. The recurrence risk of papillary thyroid carcinoma can be preoperatively predicted using miRNA-221, FN1, and CDKN2A genes.


Subject(s)
Biomarkers, Tumor , MicroRNAs , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Biopsy, Fine-Needle , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/diagnosis , Female , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/diagnosis , Male , Biomarkers, Tumor/genetics , MicroRNAs/genetics , Middle Aged , Adult , Proto-Oncogene Proteins B-raf/genetics , Mutation , Aged , Fibronectins/genetics , Fibronectins/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation, Neoplastic , Prognosis
9.
J Endocrinol ; 262(3)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38954845

ABSTRACT

Irisin is a recently discovered myokine that facilitates the browning of white adipose tissue, increases glucose uptake in skeletal muscle, and influences metabolic processes in the liver. However, its potential effects on amino acid absorption remained largely unexplored. This study aimed to elucidate the role of irisin in modulating amino acid uptake and delineate the underlying molecular mechanisms involved. To this end, juvenile tilapia were administered intraperitoneal irisin injections at 100 ng/g body weight over 8 weeks. Evaluation of various physiological parameters revealed that irisin supplementation significantly improved the specific growth rate and feed conversion efficiency while reducing feed consumption. Muscle tissue analysis revealed that irisin significantly modified the proximate composition by increasing protein content and reducing lipid levels. It also significantly raised the levels of both essential and non-essential amino acids in the muscle. Histological analysis demonstrated that irisin-stimulated muscle growth through hyperplasia rather than hypertrophy, corroborated by upregulated IGF-1 mRNA and downregulated myostatin mRNA expression. Mechanistic studies in cultured tilapia muscle cells elucidated that irisin activated integrin receptors on muscle cells, which subsequently engaged IGF-1/IGF-1R signaling. Downstream of IGF-1R activation, irisin simultaneously stimulates the ERK1/2 and PI3K/mTORC2/Akt pathways. The convergence of these pathways upregulates L-type amino acid transporter 1 expression, thereby augmenting amino acid uptake into muscle cells. In summary, irisin supplementation in tilapia leads to improved muscle growth, predominantly via hyperplasia and augmented amino acid assimilation, governed by intricate cellular signaling pathways. These findings provide valuable aquaculture applications and novel insights into muscle development.


Subject(s)
Amino Acids , Fibronectins , Insulin-Like Growth Factor I , Muscle, Skeletal , Signal Transduction , Tilapia , Animals , Insulin-Like Growth Factor I/metabolism , Tilapia/metabolism , Tilapia/growth & development , Fibronectins/metabolism , Fibronectins/genetics , Amino Acids/metabolism , Signal Transduction/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development
10.
Physiol Rep ; 12(14): e16152, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054559

ABSTRACT

Plasma fibronectin (pFN) is a hepatocyte-derived circulating extracellular matrix protein that affects cell morphology, adipogenesis, and insulin signaling of adipocytes in vitro. In this study, we show pFN accrual to adipose tissue and its contribution to tissue homeostasis in mice. Hepatocyte-specific conditional Fn1 knockout mice (Fn1-/-ALB) show a decrease in adipose tissue FN levels and enhanced insulin sensitivity of subcutaneous (inguinal), visceral (epididymal) adipose tissue on a normal diet. Diet-induced obesity model of the Fn1-/-ALB mouse showed normal weight gain and whole-body fat mass, and normal adipose tissue depot volumes and unaltered circulating leptin and adiponectin levels. However, Fn1-/-ALB adipose depots showed significant alterations in adipocyte size and gene expression profiles. The inguinal adipose tissue on a normal diet, which had alterations in fatty acid metabolism and thermogenesis suggesting browning. The presence of increased beige adipocyte markers Ucp1 and Prdm16 supported this. In the inguinal fat, the obesogenic diet resulted in downregulation of the browning markers and changes in gene expression reflecting development, morphogenesis, and mesenchymal stem cell maintenance. Epididymal adipose tissue showed alterations in developmental and stem cell gene expression on both diets. The data suggests a role for pFN in adipose tissue insulin sensitivity and cell profiles.


Subject(s)
Fibronectins , Insulin Resistance , Animals , Mice , Fibronectins/metabolism , Fibronectins/genetics , Male , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipogenesis , Mice, Knockout , Mice, Inbred C57BL , Obesity/metabolism , Obesity/genetics , Obesity/blood , Cell Differentiation , Diet, High-Fat
11.
J Med Food ; 27(8): 749-757, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39017636

ABSTRACT

The study aimed to evaluate the effects of Pereskia aculeata Miller (ora-pro-nobis [OPN]) flour on body and biochemical parameters, thermogenic activity, and molecular expression of markers in the muscle tissue of mice subjected to resistance training (RT). Twelve mice were randomly assigned to two groups (n=6 animals/group): G1: control (Control) fed a standard diet + RT and G2: experimental (OPN) fed a diet based on OPN flour + RT. The RT consisted of a 6-week program using a vertical ladder combined with a fixed weight attached to the animal. Several parameters were measured, including assessment of body composition, biochemical markers, thermogenic activity, and molecular (mRNA expression of interleukin (IL)-6, fibronectin type III domain-containing protein 5 (FNDC5), peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), nuclear respiratory factor 1 (NRF1), and mitochondrial transcription factor A (TFAM). The OPN group exhibited a decrease in body weight and visceral adiposity, higher energy expenditure, and lipid oxidation rate. In addition, it was observed an increase in muscle volume and in mRNA expression levels of IL-6, FNDC5, PGC-1α, and TFAM. These findings suggest that OPN flour could be a nutritional option to enhance performance in RT.


Subject(s)
Flour , Interleukin-6 , Muscle, Skeletal , Myokines , Resistance Training , Animals , Humans , Male , Mice , Body Composition/drug effects , Energy Metabolism , Fibronectins/metabolism , Fibronectins/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Muscle, Skeletal/metabolism , Myokines/genetics , Myokines/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Physical Conditioning, Animal , Thermogenesis/drug effects
12.
Biochem Pharmacol ; 227: 116423, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996930

ABSTRACT

The placenta experiences a low-oxygen stage during early pregnancy. Aspirin is an effective preventative treatment for preeclampsia if applied early in pregnancy. Elevation of fibronectin (FN) level has been reported to be associated with preeclampsia; however, the role of FN in the physiological hypoxic phase and whether aspirin exerts its effect on FN at this hypoxic stage remain unknown. We determined pregnancy outcomes by injecting saline or recombinant FN protein into C57BL/6 pregnant mice and one group of FN-injected mice was fed aspirin. The effects of FN, the underlying pathways on trophoblast biology, and cilia formation under hypoxia were investigated in FN-pretreated or FN-knockdown HTR-8/SVneo cells in a hypoxic chamber (0.1 % O2). Preeclampsia-like phenotypes, including blood pressure elevation and proteinuria, developed in FN-injected pregnant mice. The fetal weight of FN-injected mice was significantly lower than that of non-FN-injected mice (p < 0.005). Trophoblast FN expression was upregulated under hypoxia, which could be suppressed by aspirin treatment. FN inhibited trophoblast invasion and migration under hypoxia, and this inhibitory effect occurred through downregulating ZEB1/2, MMP 9 and the Akt and MAPK signaling pathways. Ciliogenesis of trophoblasts was stimulated under hypoxia but was inhibited by FN treatment. Aspirin was shown to reverse the FN-mediated inhibitory effect on trophoblast invasion/migration and ciliogenesis. In conclusion, FN overexpression induces preeclampsia-like symptoms and impairs fetal growth in mice. Aspirin may exert its suppressive effect on FN upregulation and FN-mediated cell function in the hypoxic stage of pregnancy and therefore provides a preventative effect on preeclampsia development.


Subject(s)
Aspirin , Fibronectins , MAP Kinase Signaling System , Mice, Inbred C57BL , Pre-Eclampsia , Proto-Oncogene Proteins c-akt , Trophoblasts , Animals , Pre-Eclampsia/metabolism , Pre-Eclampsia/prevention & control , Pre-Eclampsia/pathology , Fibronectins/metabolism , Fibronectins/genetics , Female , Pregnancy , Aspirin/pharmacology , Trophoblasts/drug effects , Trophoblasts/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Humans , Disease Models, Animal , Cilia/drug effects , Cilia/metabolism , Cilia/physiology , Phenotype , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Hypoxia/metabolism , Cell Line
13.
Microb Pathog ; 193: 106752, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880315

ABSTRACT

Arcobacter butzleri is a foodborne pathogen that mainly causes enteritis in humans, but the number of cases of bacteraemia has increased in recent years. However, there is still limited knowledge on the pathogenic mechanisms of this bacterium. To investigate how A. butzleri causes disease, single knockout mutants were constructed in the cadF, ABU_RS00335, ciaB, and flaAB genes, which might be involved in adhesion and invasion properties. These mutants and the isogenic wild-type (WT) were then tested for their ability to adhere and invade human Caco-2 and HT29-MTX cells. The adhesion and invasion of A. butzleri RM4018 strain was also visualized by a Leica CTR 6500 confocal microscope. The adhesion and invasion abilities of mutants lacking the invasion antigen CiaB or a functional flagellum were lower than those of the WTs. However, the extent of the decrease varied depending on the strain and/or cell line. Mutants lacking the fibronectin (FN)-binding protein CadF consistently exhibited reduced abilities, while the inactivation of the other studied FN-binding protein, ABU_RS00335, led to a reduction in only one of the two strains tested. Therefore, the ciaB and flaAB genes appear to be important for A. butzleri adhesion and invasion properties, while cadF appears to be indispensable.


Subject(s)
Adhesins, Bacterial , Arcobacter , Bacterial Adhesion , Flagella , Bacterial Adhesion/genetics , Humans , Arcobacter/genetics , Caco-2 Cells , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Flagella/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Knockout Techniques , HT29 Cells , Fibronectins/metabolism , Fibronectins/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Genes, Bacterial/genetics , Epithelial Cells/microbiology , Virulence/genetics
14.
Protein Eng Des Sel ; 372024 Jan 29.
Article in English | MEDLINE | ID: mdl-38836499

ABSTRACT

Protein developability is requisite for use in therapeutic, diagnostic, or industrial applications. Many developability assays are low throughput, which limits their utility to the later stages of protein discovery and evolution. Recent approaches enable experimental or computational assessment of many more variants, yet the breadth of applicability across protein families and developability metrics is uncertain. Here, three library-scale assays-on-yeast protease, split green fluorescent protein (GFP), and non-specific binding-were evaluated for their ability to predict two key developability outcomes (thermal stability and recombinant expression) for the small protein scaffolds affibody and fibronectin. The assays' predictive capabilities were assessed via both linear correlation and machine learning models trained on the library-scale assay data. The on-yeast protease assay is highly predictive of thermal stability for both scaffolds, and the split-GFP assay is informative of affibody thermal stability and expression. The library-scale data was used to map sequence-developability landscapes for affibody and fibronectin binding paratopes, which guides future design of variants and libraries.


Subject(s)
Fibronectins , Recombinant Fusion Proteins , Fibronectins/chemistry , Fibronectins/genetics , Fibronectins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Protein Engineering/methods , Peptide Library , Protein Stability , Protein Binding , Humans
15.
Crit Rev Eukaryot Gene Expr ; 34(5): 15-29, 2024.
Article in English | MEDLINE | ID: mdl-38842201

ABSTRACT

RBM15 functions as an oncogene in multi-type cancers. However, the reports on the roles of RBM15 in cervical cancer are limited. The purpose of this study was to investigate the potentials of RBM15 in cervical cancer. RT-qPCR was conducted to determine mRNA levels. Western was carried out to detect protein expression. CCK-8, colony formation and EdU assays were conducted to determine cell proliferation. Scratch and transwell assays were conducted to determine cell migration and invasion. MeRIP assay was conducted to determine N6-methyl adenosine (m6A) levels. Luciferase assay was conducted to verify the m6A sites of EZH2 and binding sites between EZH2 and promoter of FN1. ChIP assay was conducted to verify the interaction between EZH2 and FN1. The results showed that RBM15 was upregulated in cervical cancer patients and cells. Moreover, high levels of RBM15 predicted poor clinical outcomes. RBM15 knockdown inhibited the proliferation and epithelial-mesenchymal transition (EMT) of cervical cancer cells. RBM15 promoted the m6A modification of EZH2 as well as its protein translation. Additionally, EZH2 bound to the promoter of fibronectin 1 (FN1) and EZH2-FN1 axis is the cascade downstream of RBM15. Overexpressed EZH2 antagonized the effects of RBM15 knockdown and promoted the aggressiveness of cervical cancer cells. In summary, RBM15/EZH2/FN1 signaling cascade induces the proliferation and EMT of cervical cancer. Therefore, RBM15/EZH2/FN1 signaling may be a promising strategy for cervical cancer.


Subject(s)
Adenosine , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Adenosine/analogs & derivatives , Adenosine/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line, Tumor , Cell Movement , Fibronectins/metabolism , Fibronectins/genetics
16.
Vet J ; 306: 106161, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38849027

ABSTRACT

Irisin is a 112-amino acid peptide hormone that is cleaved from fibronectin type III domain-containing protein 5 (FNDC5), a type I transmembrane protein abundantly found in muscle tissue. Irisin is a putative mediator of the benefits of exercise, neuroprotection, bone growth, and cardiac health. However, few studies have focused on irisin in domestic animals. Further, whether processed irisin is detectable in domestic animal tissues remains uncertain. To address this, we determined FNDC5 mRNA and protein concentration in anatine (duck) and porcine (pig) skeletal muscle, and in equine (horse), swine, and anatine serum samples. RT-PCR analysis identified FNDC5 mRNA in all pig and duck skeletal muscle samples. An approximately 25 kDa band representing FNDC5 was detected in both pig and duck skeletal muscle. Fluorescence immunohistochemistry using a rabbit monoclonal FNDC5/irisin primary antibody and a goat polyclonal anti-rabbit secondary antibody localized FNDC5/irisin-like immunoreactivity in both the glandular and muscular regions of pig stomach. FNDC5/irisin-like immunoreactivity was also identified in horse, pig, and duck serum using a multispecies irisin ELISA. The average values of irisin-like immunoreactivity were 13.7 (duck), 15.4 (horse), and 7.0 (pig) ng/mL in samples tested. Our results support the presence of irisin precursor in several domestic animals. Processed irisin, however, was not detectable. Further studies are required to validate reliable tools to detect and quantify processed irisin in domestic animals.


Subject(s)
Ducks , Fibronectins , Muscle, Skeletal , RNA, Messenger , Animals , Horses , Fibronectins/metabolism , Fibronectins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Swine , Muscle, Skeletal/metabolism , Muscle, Skeletal/chemistry , Ducks/metabolism
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 303-310, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710514

ABSTRACT

Objective To clarify the relationship between astrocyte activation patterns and disease progression in epidemic encephalitis B (Japanese encephalitis). Methods First, a mouse model of epidemic encephalitis B was constructed by foot-pad injection of Japanese encephalitis virus (JEV), and the expression of viral protein NS3 in different brain regions was detected by immunofluorescence assay (IFA). Next, IFA, RNA sequencing (RNA-seq) and real-time quantitative PCR (qRT-PCR) were used to clarify the changes in the astrocyte activation patterns at different stages of epidemic encephalitis B. Finally, intracerebroventricular administration of irisin was conducted to regulate the proportion of activation in complement C3-positive A1 astrocytes and S100A10-positive A2 astrocytes, investigating whether it could improve the body mass, behavioral scores, and brain tissue damage in a mouse model. Results NS3 protein was detected by IFA predominantly in the M1/M2 region of the motor cortex and the hippocampus. The number and volume of GFAP-positive astrocytes significantly increased in JEV-infected brain regions, in which the expression of multiple genes associated with A1/A2 astrocyte activation was significantly enhanced. Although intracerebroventricular or intraperitoneal injection of irisin did not improve the prognosis of epidemic encephalitis B, it inhibited the activation of A1 astrocytes and ameliorate neuroinflammation. Conclusion Neurons in the M1/M2 motor cortex and hippocampus are susceptible to JEV infection, in which the abnormal astrocyte activation contributes to the neuroinflammatory injury. Irisin administration may restrain A1 astrocyte activation and alleviate neuroinflammation following JEV infection.


Subject(s)
Astrocytes , Disease Models, Animal , Disease Progression , Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Astrocytes/metabolism , Astrocytes/virology , Mice , Encephalitis, Japanese/immunology , Encephalitis Virus, Japanese/physiology , Brain/metabolism , Brain/virology , Brain/pathology , Male , Fibronectins/metabolism , Fibronectins/genetics
18.
Nat Commun ; 15(1): 3732, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702309

ABSTRACT

Immunotherapy with chimeric antigen receptor T cells for pediatric solid and brain tumors is constrained by available targetable antigens. Cancer-specific exons present a promising reservoir of targets; however, these have not been explored and validated systematically in a pan-cancer fashion. To identify cancer specific exon targets, here we analyze 1532 RNA-seq datasets from 16 types of pediatric solid and brain tumors for comparison with normal tissues using a newly developed workflow. We find 2933 exons in 157 genes encoding proteins of the surfaceome or matrisome with high cancer specificity either at the gene (n = 148) or the alternatively spliced isoform (n = 9) level. Expression of selected alternatively spliced targets, including the EDB domain of fibronectin 1, and gene targets, such as COL11A1, are validated in pediatric patient derived xenograft tumors. We generate T cells expressing chimeric antigen receptors specific for the EDB domain or COL11A1 and demonstrate that these have antitumor activity. The full target list, explorable via an interactive web portal ( https://cseminer.stjude.org/ ), provides a rich resource for developing immunotherapy of pediatric solid and brain tumors using gene or AS targets with high expression specificity in cancer.


Subject(s)
Brain Neoplasms , Exons , Receptors, Chimeric Antigen , Humans , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/genetics , Animals , Exons/genetics , Child , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Mice , Immunotherapy/methods , Alternative Splicing , Fibronectins/genetics , Fibronectins/metabolism , Fibronectins/immunology , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , RNA-Seq , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Immunotherapy, Adoptive/methods
19.
Nat Commun ; 15(1): 4546, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806494

ABSTRACT

Asthma has striking disparities across ancestral groups, but the molecular underpinning of these differences is poorly understood and minimally studied. A goal of the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA) is to understand multi-omic signatures of asthma focusing on populations of African ancestry. RNASeq and DNA methylation data are generated from nasal epithelium including cases (current asthma, N = 253) and controls (never-asthma, N = 283) from 7 different geographic sites to identify differentially expressed genes (DEGs) and gene networks. We identify 389 DEGs; the top DEG, FN1, was downregulated in cases (q = 3.26 × 10-9) and encodes fibronectin which plays a role in wound healing. The top three gene expression modules implicate networks related to immune response (CEACAM5; p = 9.62 × 10-16 and CPA3; p = 2.39 × 10-14) and wound healing (FN1; p = 7.63 × 10-9). Multi-omic analysis identifies FKBP5, a co-chaperone of glucocorticoid receptor signaling known to be involved in drug response in asthma, where the association between nasal epithelium gene expression is likely regulated by methylation and is associated with increased use of inhaled corticosteroids. This work reveals molecular dysregulation on three axes - increased Th2 inflammation, decreased capacity for wound healing, and impaired drug response - that may play a critical role in asthma within the African Diaspora.


Subject(s)
Asthma , Black People , DNA Methylation , Nasal Mucosa , Tacrolimus Binding Proteins , Humans , Asthma/genetics , Asthma/metabolism , Nasal Mucosa/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Female , Male , Black People/genetics , Adult , Gene Regulatory Networks , Fibronectins/metabolism , Fibronectins/genetics , Case-Control Studies , Gene Expression Regulation , Middle Aged , Multiomics
20.
Acta Physiol (Oxf) ; 240(7): e14163, 2024 07.
Article in English | MEDLINE | ID: mdl-38752665

ABSTRACT

AIM: To reveal the contribution of Irisin in the beneficial effects of resistance exercise on myocardial fibrosis (MF) and cardiac function in the mice with myocardial infarction (MI). METHODS: The MI model was built by ligating the left anterior descending coronary artery in Fndc5 knockout mice (Fndc5-/-). Resistance exercise was started one week after surgery and continued for four weeks. In addition, H2O2, AICAR, recombinant human Irisin protein (rhIRISIN), and Sirt1 shRNA lentivirus (LV-Sirt1 shRNA) were used to intervene primary isolated cardiac fibroblasts (CFs). MF was observed through Masson staining, and apoptosis was assessed using TUNEL staining. MDA and T-SOD contents were detected by biochemical kits. The expression of proteins and genes was detected by Western blotting and RT-qPCR. RESULTS: Resistance exercise increased Fndc5 mRNA level, inhibited the activation of TGFß1-TGFßR2-Smad2/3 pathway, activated AMPK-Sirt1 pathway, reduced the levels of oxidative stress, apoptosis, and MF in the infarcted heart, and promoted cardiac function. However, Fndc5 knockout attenuated the protective effects of resistance exercise on the MI heart. Results of the in vitro experiments showed that AICAR and rhIRISIN intervention activated the AMPK-Sirt1 pathway and inactivated the TGFß1-Smad2/3 pathway, and promoted apoptosis in H2O2-treated CFs. Notably, these effects of rhIRISIN intervention, except for the TGFßR2 expression, were attenuated by LV-Sirt1 shRNA. CONCLUSION: Resistance exercise upregulates Fndc5 expression, activates AMPK-Sirt1 pathway, inhibits the activation of TGFß1-Smad2/3 pathway, attenuates MF, and promotes cardiac function after MI.


Subject(s)
AMP-Activated Protein Kinases , Fibronectins , Fibrosis , Mice, Knockout , Myocardial Infarction , Sirtuin 1 , Transforming Growth Factor beta1 , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Sirtuin 1/metabolism , Sirtuin 1/genetics , Fibronectins/metabolism , Fibronectins/genetics , Mice , Fibrosis/metabolism , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Transforming Growth Factor beta1/metabolism , Smad2 Protein/metabolism , Up-Regulation , Resistance Training , Male , Myocardium/metabolism , Myocardium/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Physical Conditioning, Animal/physiology , Mice, Inbred C57BL , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL