Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.328
Filter
1.
Am J Physiol Cell Physiol ; 326(5): C1482-C1493, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38525537

ABSTRACT

Corneal fibroblasts maintain homeostasis of the corneal stroma by mediating the synthesis and degradation of extracellular collagen, and these actions are promoted by transforming growth factor-ß (TGF-ß) and interleukin-1ß (IL-1ß), respectively. The cornea is densely innervated with sensory nerve fibers that are not only responsible for sensation but also required for physiological processes such as tear secretion and wound healing. Loss or dysfunction of corneal nerves thus impairs corneal epithelial wound healing and can lead to neurotrophic keratopathy. The sensory neurotransmitter substance P (SP) promotes corneal epithelial wound healing by enhancing the stimulatory effects of growth factors and fibronectin. We have now investigated the role of SP in collagen metabolism mediated by human corneal fibroblasts in culture. Although SP alone had no effect on collagen synthesis or degradation by these cells, it promoted the stimulatory effect of TGF-ß on collagen type I synthesis without affecting that of IL-1ß on the expression of matrix metalloproteinase-1. This effect of SP on TGF-ß-induced collagen synthesis was accompanied by activation of p38 mitogen-activated protein kinase (MAPK) signaling and was attenuated by pharmacological inhibition of p38 or of the neurokinin-1 receptor. Our results thus implicate SP as a modulator of TGF-ß-induced collagen type I synthesis by human corneal fibroblasts, and they suggest that loss of this function may contribute to the development of neurotrophic keratopathy.NEW & NOTEWORTHY This study investigates the role of substance P (SP) in collagen metabolism mediated by human corneal fibroblasts in culture. We found that, although SP alone had no effect on collagen synthesis or degradation by corneal fibroblasts, it promoted the stimulatory effect of transforming growth factor-ß on collagen type I synthesis without affecting that of interleukin-1ß on the expression of matrix metalloproteinase-1.


Subject(s)
Fibroblasts , Interleukin-1beta , Substance P , Transforming Growth Factor beta , p38 Mitogen-Activated Protein Kinases , Humans , Substance P/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Transforming Growth Factor beta/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Cells, Cultured , Interleukin-1beta/metabolism , Collagen Type I/metabolism , Collagen Type I/biosynthesis , Receptors, Neurokinin-1/metabolism , Cornea/metabolism , Cornea/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Collagen/metabolism , Collagen/biosynthesis , Signal Transduction/drug effects , Corneal Stroma/metabolism , Corneal Stroma/drug effects , Corneal Keratocytes/metabolism , Corneal Keratocytes/drug effects
2.
Toxicol Appl Pharmacol ; 491: 117078, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214171

ABSTRACT

RUNX1 with CBFß functions as an activator or repressor of critical mediators regulating cellular function. The aims of this study were to clarify the role of RUNX1 on regulating TGF-ß1-induced COL1 synthesis and the mechanism of calcipotriol (Cal) on antagonizing COL1 synthesis in PSCs. RT-qPCR and Western Blot for determining the mRNAs and proteins of RUNX1 and COL1A1/1A2 in rat PSC line (RP-2 cell). Luciferase activities driven by RUNX1 or COL1A1 or COL1A2 promoter, co-immunoprecipitation and immunoblotting for pSmad3/RUNX1 or CBFß/RUNX1, and knockdown or upregulation of Smad3 and RUNX1 were used. RUNX1 production was regulated by TGF-ß1/pSmad3 signaling pathway in RP-2 cells. RUNX1 formed a coactivator with CBFß in TGF-ß1-treated RP-2 cells to regulate the transcriptions of COL1A1/1A2 mRNAs under a fashion of pSmad3/RUNX1/CBFß complex. However, Cal effectively abrogated the levels of COL1A1/1A2 transcripts in TGF-ß1-treated RP-2 cells by downregulating RUNX1 production and hindering the formation of pSmad3/RUNX1/CBFß complexes. This study suggests that RUNX1 may be a promising antifibrotic target for the treatment of chronic pancreatitis.


Subject(s)
Calcitriol , Collagen Type I , Core Binding Factor Alpha 2 Subunit , Down-Regulation , Pancreatic Stellate Cells , Smad3 Protein , Transforming Growth Factor beta1 , Animals , Calcitriol/pharmacology , Calcitriol/analogs & derivatives , Transforming Growth Factor beta1/metabolism , Smad3 Protein/metabolism , Rats , Down-Regulation/drug effects , Collagen Type I/metabolism , Collagen Type I/biosynthesis , Collagen Type I/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Cell Line , Signal Transduction/drug effects
3.
J Biol Chem ; 298(9): 102246, 2022 09.
Article in English | MEDLINE | ID: mdl-35835217

ABSTRACT

Proximal tubular epithelial cells respond to transforming growth factor ß (TGFß) to synthesize collagen I (α2) during renal fibrosis. The oncoprotein DJ-1 has previously been shown to promote tumorigenesis and prevent apoptosis of dopaminergic neurons; however, its role in fibrosis signaling is unclear. Here, we show TGFß-stimulation increased expression of DJ-1, which promoted noncanonical mTORC1 and mTORC2 activities. We show DJ-1 augmented the phosphorylation/activation of PKCßII, a direct substrate of mTORC2. In addition, coimmunoprecipitation experiments revealed association of DJ-1 with Raptor and Rictor, exclusive subunits of mTORC1 and mTORC2, respectively, as well as with mTOR kinase. Interestingly, siRNAs against DJ-1 blocked TGFß-stimulated expression of collagen I (α2), while expression of DJ-1 increased expression of this protein. In addition, expression of dominant negative PKCßII and siRNAs against PKCßII significantly inhibited TGFß-induced collagen I (α2) expression. In fact, constitutively active PKCßII abrogated the effect of siRNAs against DJ-1, suggesting a role of PKCßII downstream of this oncoprotein. Moreover, we demonstrate expression of collagen I (α2) stimulated by DJ-1 and its target PKCßII is dependent on the transcription factor hypoxia-inducible factor 1α (Hif1α). Finally, we show in the renal cortex of diabetic rats that increased TGFß was associated with enhanced expression of DJ-1 and activation of mTOR and PKCßII, concomitant with increased Hif1α and collagen I (α2). Overall, we identified that DJ-1 affects TGFß-induced expression of collagen I (α2) via an mTOR-, PKCßII-, and Hif1α-dependent mechanism to regulate renal fibrosis.


Subject(s)
Collagen Type I , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Oncogene Proteins , Protein Deglycase DJ-1 , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Protein Kinase C beta/metabolism , RNA, Small Interfering/metabolism , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
4.
J Biol Chem ; 296: 100027, 2021.
Article in English | MEDLINE | ID: mdl-33154166

ABSTRACT

Osteogenesis imperfecta (OI) is a heritable brittle bone disease mainly caused by mutations in the two type I collagen genes. Collagen synthesis is a complex process including trimer formation, glycosylation, secretion, extracellular matrix (ECM) formation, and mineralization. Using OI patient-derived fibroblasts and induced pluripotent stem cells (iPSCs), we investigated the effect of 4-phenylbutyric acid (4-PBA) on collagen synthesis to test its potential as a new treatment for OI. Endoplasmic reticulum (ER) retention of type I collagen was observed by immunofluorescence staining in OI patient-derived fibroblasts with glycine substitution and exon skipping mutations. Liquid chromatography-mass spectrometry analysis revealed excessive glycosylation of secreted type I collagen at the specific sites in OI cells. The misfolding of the type I collagen triple helix in the ECM was demonstrated by the incorporation of heat-dissociated collagen hybridizing peptide in OI cells. Type I collagen was produced excessively by OI fibroblasts with a glycine mutation, but this excessive production was normalized when OI fibroblasts were cultured on control fibroblast-derived ECM. We also found that mineralization was impaired in osteoblasts differentiated from OI iPSCs. In summary, treatment with 4-PBA normalizes the excessive production of type I collagen, reduces ER retention, partially improves misfolding of the type I collagen helix in ECM, and improves osteoblast mineralization. Thus, 4-PBA may improve not only ER retention, but also type I collagen synthesis and mineralization in human cells from OI patients.


Subject(s)
Calcification, Physiologic/drug effects , Induced Pluripotent Stem Cells/drug effects , Osteoblasts/drug effects , Osteogenesis Imperfecta/pathology , Phenylbutyrates/pharmacology , Cell Differentiation , Child, Preschool , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Humans , Mutation , Osteoblasts/cytology , Osteogenesis Imperfecta/metabolism , Protein Folding
5.
Int Urogynecol J ; 33(7): 1803-1812, 2022 07.
Article in English | MEDLINE | ID: mdl-35596801

ABSTRACT

INTRODUCTION AND HYPOTHESIS: This study was aimed at identifying the difference in collagen type-1 expression in women with and without pelvic organ prolapse (POP). METHODS: A systematic review and meta-analysis was carried out women with and without pelvic organ prolapse. This meta-analysis was conducted on research articles describing the evaluation of collagen type-1 expression between patients with and without POP. The articles were obtained from PubMed, EBSCO, and ProQuest, and were published between January 2000 and June 2021. Pooled mean difference (MD) and pooled odds ratio (OR) were calculated using fixed effect models. Review Manager (RevMan 5.4) was used to analyze the data. The main outcome measures were pooled MD and pooled OR of collagen type-1 expression in patients with and without POP. RESULTS: A total of seven case-control studies were included in the meta-analysis using the effect size of the MD and two case-control studies were included in the meta-analysis using the effect size of the OR. A total of 247 POP cases and 132 non-POP cases were identified from the studies. Our study indicated that patients with POP had a lower level of collagen type-1 expression than non-POP patients (MD = -6.77; 95% CI: -8.37, -5.17, p < 0.00001). Patients with low expression of collagen type-1 in pelvic support tissue are at a more than 3 times higher risk of suffering from pelvic organ prolapse (OR = 3.23, 95% CI: 1.52 to 6.87, p = 0.002). CONCLUSION: The results of this study showed that patients with pelvic organ prolapse have lower expression of collagen type-1 than nonpelvic organ prolapse patients.


Subject(s)
Collagen Type I , Pelvic Organ Prolapse , Collagen Type I/biosynthesis , Collagen Type I/metabolism , Female , Humans , Pelvic Organ Prolapse/metabolism
6.
PLoS Genet ; 15(6): e1008196, 2019 06.
Article in English | MEDLINE | ID: mdl-31173582

ABSTRACT

Covalent intermolecular cross-linking of collagen is essential for tissue stability. Recent studies have demonstrated that cyclophilin B (CypB), an endoplasmic reticulum (ER)-resident peptidyl-prolyl cis-trans isomerase, modulates lysine (Lys) hydroxylation of type I collagen impacting cross-linking chemistry. However, the extent of modulation, the molecular mechanism and the functional outcome in tissues are not well understood. Here, we report that, in CypB null (KO) mouse skin, two unusual collagen cross-links lacking Lys hydroxylation are formed while neither was detected in wild type (WT) or heterozygous (Het) mice. Mass spectrometric analysis of type I collagen showed that none of the telopeptidyl Lys was hydroxylated in KO or WT/Het mice. Hydroxylation of the helical cross-linking Lys residues was almost complete in WT/Het but was markedly diminished in KO. Lys hydroxylation at other sites was also lower in KO but to a lesser extent. A key glycosylation site, α1(I) Lys-87, was underglycosylated while other sites were mostly overglycosylated in KO. Despite these findings, lysyl hydroxylases and glycosyltransferase 25 domain 1 levels were significantly higher in KO than WT/Het. However, the components of ER chaperone complex that positively or negatively regulates lysyl hydroxylase activities were severely reduced or slightly increased, respectively, in KO. The atomic force microscopy-based nanoindentation modulus were significantly lower in KO skin than WT. These data demonstrate that CypB deficiency profoundly affects Lys post-translational modifications of collagen likely by modulating LH chaperone complexes. Together, our study underscores the critical role of CypB in Lys modifications of collagen, cross-linking and mechanical properties of skin.


Subject(s)
Cyclophilins/chemistry , Lysine/chemistry , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/chemistry , Skin/enzymology , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Cyclophilins/genetics , Cyclophilins/ultrastructure , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/enzymology , Glycosylation , Heterozygote , Hydroxylation , Lysine/genetics , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Atomic Force , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Protein Processing, Post-Translational/genetics , Skin/chemistry
7.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L377-L391, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34105356

ABSTRACT

Genome-wide association studies have shown that a gene variant in the Family with sequence similarity 13, member A (FAM13A) is strongly associated with reduced lung function and the appearance of respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD). A key player in smoking-induced tissue injury and airway remodeling is the transforming growth factor-ß1 (TGF-ß1). To determine the role of FAM13A in TGF-ß1 signaling, FAM13A-/- airway epithelial cells were generated using CRISPR-Cas9, whereas overexpression of FAM13A was achieved using lipid nanoparticles. Wild-type (WT) and FAM13A-/- cells were treated with TGF-ß1, followed by gene and/or protein expression analyses. FAM13A-/- cells augmented TGF-ß1-induced increase in collagen type 1 (COL1A1), matrix metalloproteinase 2 (MMP2), expression compared with WT cells. This effect was mediated by an increase in ß-catenin (CTNNB1) expression in FAM13A-/- cells compared with WT cells after TGF-ß1 treatment. FAM13A overexpression was partially protective from TGF-ß1-induced COL1A1 expression. Finally, we showed that airway epithelial-specific FAM13A protein expression is significantly increased in patients with severe COPD compared with control nonsmokers, and negatively correlated with lung function. In contrast, ß-catenin (CTNNB1), which has previously been linked to be regulated by FAM13A, is decreased in the airway epithelium of smokers with COPD compared with non-COPD subjects. Together, our data showed that FAM13A may be protective from TGF-ß1-induced fibrotic response in the airway epithelium via sequestering CTNNB1 from its regulation on downstream targets. Therapeutic increase in FAM13A expression in the airway epithelium of smokers at risk for COPD, and those with mild COPD, may reduce the extent of airway tissue remodeling.


Subject(s)
Airway Remodeling , GTPase-Activating Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism , Smoking/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Aged , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology , Smoking/genetics , Smoking/pathology , Transforming Growth Factor beta1/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
8.
Hum Mol Genet ; 28(20): 3431-3442, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31411676

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the gene encoding dystrophin. Prior work has shown that DMD progression can vary, depending on the genetic makeup of the patient. Several modifier alleles have been identified including LTBP4 and SPP1. We previously showed that Spp1 exacerbates the DMD phenotype in the mdx mouse model by promoting fibrosis and by skewing macrophage polarization. Here, we studied the mechanisms involved in Spp1's promotion of fibrosis by using both isolated fibroblasts and genetically modified mice. We found that Spp1 upregulates collagen expression in mdx fibroblasts by enhancing TGFß signaling. Spp1's effects on TGFß signaling are through induction of MMP9 expression. MMP9 is a protease that can release active TGFß ligand from its latent complex. In support for activation of this pathway in our model, we showed that treatment of mdx fibroblasts with MMP9 inhibitor led to accumulation of the TGFß latent complex, decreased levels of active TGFß and reduced collagen expression. Correspondingly, we found reduced active TGFß in Spp1-/-mdxB10 and Mmp9-/-mdxB10 muscles in vivo. Taken together with previous observations of reduced fibrosis in both models, these data suggest that Spp1 acts upstream of TGFß to promote fibrosis in mdx muscles. We found that in the context of constitutively upregulated TGFß signaling (such as in the mdxD2 model), ablation of Spp1 has very little effect on fibrosis. Finally, we performed proof-of-concept studies showing that postnatal pharmacological inhibition of Spp1 reduces fibrosis and improves muscle function in mdx mice.


Subject(s)
Fibrosis/genetics , Muscular Dystrophy, Duchenne/metabolism , Osteopontin/genetics , Transforming Growth Factor beta/metabolism , Animals , Collagen Type I/biosynthesis , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Osteopontin/metabolism , Primary Cell Culture , Regeneration/genetics , Signal Transduction , Transforming Growth Factor beta/genetics
9.
Hum Genet ; 140(8): 1121-1141, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34169326

ABSTRACT

Osteogenesis Imperfecta (OI) comprises a heterogeneous group of patients who share bone fragility and deformities as the main characteristics, albeit with different degrees of severity. Phenotypic variation also exists in other connective tissue aspects of the disease, complicating disease classification and disease course prediction. Although collagen type I defects are long established as the primary cause of the bone pathology, we are still far from comprehending the complete mechanism. In the last years, the advent of next generation sequencing has triggered the discovery of many new genetic causes for OI, helping to draw its molecular landscape. It has become clear that, in addition to collagen type I genes, OI can be caused by multiple proteins connected to different parts of collagen biosynthesis. The production of collagen entails a complex process, starting from the production of the collagen Iα1 and collagen Iα2 chains in the endoplasmic reticulum, during and after which procollagen is subjected to a plethora of posttranslational modifications by chaperones. After reaching the Golgi organelle, procollagen is destined to the extracellular matrix where it forms collagen fibrils. Recently discovered mutations in components of the retrograde transport of chaperones highlight its emerging role as critical contributor of OI development. This review offers an overview of collagen regulation in the context of recent gene discoveries, emphasizing the significance of transport disruptions in the OI mechanism. We aim to motivate exploration of skeletal fragility in OI from the perspective of these pathways to identify regulatory points which can hint to therapeutic targets.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/biosynthesis , Osteoblasts/metabolism , Osteogenesis Imperfecta/metabolism , Procollagen/biosynthesis , Protein Processing, Post-Translational , Bone and Bones/pathology , Collagen Type I/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , High-Throughput Nucleotide Sequencing , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Osteoblasts/pathology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Procollagen/genetics , Protein Biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Transport , Severity of Illness Index
10.
Histochem Cell Biol ; 155(4): 503-512, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33398434

ABSTRACT

Hypertension leads to structural remodeling of cerebral blood vessels, which has been implicated in the pathophysiology of cerebrovascular diseases. The remodeling and progression of arteriolosclerosis under hypertension involve fibrosis along with the production of type I collagen around cerebral arterioles. However, the source and regulatory mechanisms of this collagen production remain elusive. In this study, we examined if perivascular macrophages (PVMs) are involved in collagen production around cerebral small vessels in hypertensive SHRSP/Izm rats. Immunoreactivity for type I collagen around cerebral small vessels in 12-week-old hypertensive rats tended to higher than those in 4-week-old hypertensive and 12-week-old control rats. In ultrastructural analyses using transmission electron microscopy, the substantial deposition of collagen fibers could be observed in the intercellular spaces around PVMs near the arterioles of rats with prolonged hypertension. In situ hybridization analyses revealed that cells positive for mRNA of Col1a1, which comprises type I collagen, were observed near cerebral small vessels. The Col1a1-positive cells around cerebral small vessels were colocalized with immunoreactivity for CD206, a marker for PVMs, but not with those for glial fibrillary acidic protein or desmin, markers for other perivascular cells such as astrocytes and vascular smooth muscle cells. These results demonstrated that enhanced production of type I collagen is observed around cerebral small vessels in rats with prolonged hypertension and Col1a1 is expressed by PVMs, and support the concept that PVMs are involved in collagen production and vascular fibrosis under hypertensive conditions.


Subject(s)
Cerebral Arteries/metabolism , Collagen Type I/biosynthesis , Hypertension/metabolism , Macrophages/metabolism , Animals , Disease Models, Animal , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY
11.
Br J Nutr ; 126(3): 321-336, 2021 08 14.
Article in English | MEDLINE | ID: mdl-32718370

ABSTRACT

In the current research, a 60-d experiment was conducted with the purpose of exploring the impacts of methionine (Met) on growth performance, muscle nutritive deposition, muscle fibre growth and type I collagen synthesis as well as the related signalling pathway. Six diets (iso-nitrogenous) differing in Met concentrations (2·54, 4·85, 7·43, 10·12, 12·40 and 15·11 g/kg diets) were fed to 540 grass carp (178·47 (SD 0·36) g). Results showed (P < 0·05) that compared with Met deficiency, optimal level of dietary Met (1) increased feed intake, feed efficiency, specific growth rate and percentage weight gain (PWG); (2) increased fish muscle protein, lipid and free amino acid contents and improved fish muscle fatty acid profile as well as increased protein content in part associated with the target of rapamycin complex 1 (TORC1)/S6K1 signalling pathway; (3) increased the frequency distribution of muscle fibre with >50 µm of diameter; (4) increased type I collagen synthesis partly related to the transforming growth factor-ß1/Smads and CK2/TORC1 signalling pathways. In conclusion, dietary Met improved muscle growth, which might be due to the regulation of muscle nutritive deposition, muscle fibre growth and type I collagen synthesis-related signal molecules. Finally, according to PWG and muscle collagen content, the Met requirements for on-growing grass carp (178-626 g) were estimated to be 9·56 g/kg diet (33·26 g/kg protein of diet) and 9·28 g/kg diet (32·29 g/kg of dietary protein), respectively.


Subject(s)
Carps , Collagen Type I/biosynthesis , Methionine/administration & dosage , Muscle Fibers, Skeletal/physiology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Carps/growth & development , Diet/veterinary , Dietary Supplements , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction
12.
Med Sci Monit ; 27: e927577, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33386384

ABSTRACT

BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a serious irreversible lung disease. The mechanism of immune checkpoint in idiopathic pulmonary fibrosis is still unknown. MATERIAL AND METHODS First, the expression levels of PD-1/PD-L1 on the surface of CD4+ T cells and the proportion of Treg cells in IPF or controls were detected by flow cytometry. Then, expression of TGF-ß in blood samples was detected with ELISA. Moreover, a co-culture system was composed of fibroblasts stimulated by TGF-ß and CD4+ T cells from healthy people. The proportions of Treg cells and PD-1 in the co-culture system were detected. In addition, we detected the proportion of Treg cells and the level of collagen-1 after adding PD-1 or PD-L1 protein antibody blocker to the co-culture system. RESULTS Flow cytometry revealed the upregulated expression of PD-1/PD-L1 in CD4+ T cells of IPF patients. PD-1 appears to inhibit the differentiation of CD4+ T cells into Treg cells. Co-culture of myofibroblasts and CD4+ T cells induced the generation of collagen-1 and reduced the proliferation of CD4+ T cells. When PD-1 was blocked, the inhibition of Treg cell differentiation was reversed, accompanied by decreased collagen-1 production. CONCLUSIONS This work identified the molecular mechanism of PD-1 in patients with IPF. It may provide a new perspective on the therapeutic effect of PD-1.


Subject(s)
B7-H1 Antigen/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/immunology , Aged , Cell Differentiation , Cell Proliferation , Collagen Type I/biosynthesis , Female , Humans , Male , Myofibroblasts/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism
13.
Int J Mol Sci ; 22(9)2021 May 05.
Article in English | MEDLINE | ID: mdl-34063118

ABSTRACT

Mesenchymal stem cells (MSCs) have been widely used in therapeutic applications for many decades. However, more and more evidence suggests that factors such as the site of origin and pre-implantation treatment have a crucial impact on the result. This study investigates the role of freshly isolated MSCs in the lacrimal gland after allogeneic transplantation. For this purpose, MSCs from transgenic GFP mice were isolated and transplanted into allogeneic and syngeneic recipients. While the syngeneic MSCs maintained a spherical shape, allogeneic MSCs engrafted into the tissue as spindle-shaped cells in the interstitial stroma. Furthermore, the MSCs produced collagen type I in more than 85% to 95% of the detected GFP+ MSCs in the recipients of both models, supposedly contributing to pathogenic fibrosis in allogeneic recipients compared to syngeneic models. These findings indicate that allogeneic MSCs act completely differently from syngeneic MSCs, highlighting the importance of understanding the exact mechanisms behind MSCs.


Subject(s)
Bone Marrow Transplantation , Collagen Type I/biosynthesis , Mesenchymal Stem Cells/metabolism , Animals , Lacrimal Apparatus/cytology , Mesenchymal Stem Cells/cytology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Transplantation, Homologous , Transplantation, Isogeneic
14.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830270

ABSTRACT

The extracellular matrix of the bladder consists mostly of type I and III collagen, which are required during loading. During bladder injury, there is an accumulation of collagen that impairs bladder function. Little is known about the genes that regulate production of collagens in the bladder. We demonstrate that the transcription factor Odd-skipped related 1 (Osr1) is expressed in the bladder mesenchyme and epithelium at the onset of development. As development proceeds, Osr1 is mainly expressed in mesenchymal progenitors and their derivatives. We hypothesized that Osr1 regulates mesenchymal cell differentiation and production of collagens in the bladder. To test this hypothesis, we examined newborn and adult mice heterozygous for Osr1, Osr1+/-. The bladders of newborn Osr1+/- mice had a decrease in collagen I by western blot analysis and a global decrease in collagens using Sirius red staining. There was also a decrease in the cellularity of the lamina propria, where most collagen is synthesized. This was not due to decreased proliferation or increased apoptosis in this cell population. Surprisingly, the bladders of adult Osr1+/- mice had an increase in collagen that was associated with abnormal bladder function; they also had a decrease in bladder capacity and voided more frequently. The results suggest that Osr1 is important for the differentiation of mesenchymal cells that give rise to collagen-producing cells.


Subject(s)
Collagen Type I/biosynthesis , Mesenchymal Stem Cells/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Urinary Bladder/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Organogenesis/genetics , Transcription Factors/genetics
15.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071003

ABSTRACT

Superoxide dismutase (SOD) is a major antioxidant enzyme for superoxide removal, and cytoplasmic SOD (SOD1) is expressed as a predominant isoform in all cells. We previously reported that renal SOD1 deficiency accelerates the progression of diabetic nephropathy (DN) via increasing renal oxidative stress. To evaluate whether the degree of SOD1 expression determines regeneration capacity and sarcopenic phenotypes of skeletal muscles under incipient and advanced DN conditions, we investigated the alterations of SOD1 expression, oxidative stress marker, inflammation, fibrosis, and regeneration capacity in cardiotoxin (CTX)-injured tibialis anterior (TA) muscles of two Akita diabetic mouse models with different susceptibility to DN, DN-resistant C57BL/6-Ins2Akita and DN-prone KK/Ta-Ins2Akita mice. Here, we report that KK/Ta-Ins2Akita mice, but not C57BL/6-Ins2Akita mice, exhibit delayed muscle regeneration after CTX injection, as demonstrated by the finding indicating significantly smaller average cross-sectional areas of regenerating TA muscle myofibers relative to KK/Ta-wild-type mice. Furthermore, we observed markedly reduced SOD1 expression in CTX-injected TA muscles of KK/Ta-Ins2Akita mice, but not C57BL/6-Ins2Akita mice, along with increased inflammatory cell infiltration, prominent fibrosis and superoxide overproduction. Our study provides the first evidence that SOD1 reduction and the following superoxide overproduction delay skeletal muscle regeneration through induction of overt inflammation and fibrosis in a mouse model of progressive DN.


Subject(s)
Diabetic Nephropathies/complications , Muscle, Skeletal/drug effects , Nerve Regeneration/drug effects , Sarcopenia/etiology , Superoxide Dismutase-1/drug effects , Animals , Cardiotoxins/toxicity , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Progression , Enzyme Induction/drug effects , Fibrosis , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Glomerular Mesangium/pathology , Inflammation , Insulin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Oxidative Stress/drug effects , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/physiology , Superoxides/metabolism
16.
J Biol Chem ; 294(20): 8218-8226, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30952701

ABSTRACT

Excessive activation of the renin-angiotensin system (RAS) in diabetic cardiomyopathy (DCM) provokes a series of structural and functional abnormalities, and causes ventricular remodeling and heart failure in diabetes. (Pro)renin receptor (PRR) is a component of the RAS and has been reported to be up-regulated in some cardiovascular diseases. Furthermore, PRR blockade in some cardiovascular diseases, such as myocardial infarction and hypertension, has been demonstrated to reverse their pathogenesis. However, there have been few studies about the function of PRR in the pathogenesis of DCM. In this study, we hypothesized that PRR is involved in the pathogenesis of DCM and mediates myocardial injury in DCM. To explore the role of PRR in DCM, we evaluated the effects of PRR overexpression and knockdown on the DCM phenotype in vivo and in vitro The results show that PRR overexpression exacerbates myocardial injury and the inflammatory response in rats with DCM. Conversely, PRR knockdown alleviates myocardial fibrosis, apoptosis, and the inflammatory response, reversing the cardiac dysfunction in rats with DCM. In cell experiments, PRR overexpression also up-regulated the protein expression of collagen I and fibronectin, aggravated the inflammatory response, and increased the production of reactive oxygen species, whereas PRR knockdown had the opposite effect. Thus, PRR mediates myocardial injury, apoptosis, and the inflammatory response, likely through a PRR/extracellular signal-regulated kinase/reactive oxygen species pathway.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , MAP Kinase Signaling System , Myocardium/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System , Animals , Collagen Type I/biosynthesis , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/pathology , Fibronectins/biosynthesis , Fibrosis , Inflammation/metabolism , Inflammation/pathology , Male , Myocardium/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Prorenin Receptor
17.
Am J Physiol Gastrointest Liver Physiol ; 318(2): G336-G351, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31905025

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease, characterized by excess fat accumulation (steatosis). Nonalcoholic steatohepatitis (NASH) develops in 15-20% of NAFLD patients and frequently progresses to liver fibrosis and cirrhosis. We aimed to develop an ex vivo model of inflammation and fibrosis in steatotic murine precision-cut liver slices (PCLS). NASH was induced in C57Bl/6 mice on an amylin and choline-deficient l-amino acid-defined (CDAA) diet. PCLS were prepared from steatohepatitic (sPCLS) and control (cPCLS) livers and cultured for 48 h with LPS, TGFß1, or elafibranor. Additionally, C57Bl/6 mice were placed on CDAA diet for 12 wk to receive elafibranor or vehicle from weeks 7 to 12. Effects were assessed by transcriptome analysis and procollagen Iα1 protein production. The diets induced features of human NASH. Upon culture, all PCLS showed an increased gene expression of fibrosis- and inflammation-related markers but decreased lipid metabolism markers. LPS and TGFß1 affected sPCLS more pronouncedly than cPCLS. TGFß1 increased procollagen Iα1 solely in cPCLS. Elafibranor ameliorated fibrosis and inflammation in vivo but not ex vivo, where it only increased the expression of genes modulated by PPARα. sPCLS culture induced inflammation-, fibrosis-, and lipid metabolism-related transcripts, explained by spontaneous activation. sPCLS remained responsive to proinflammatory and profibrotic stimuli on gene expression. We consider that PCLS represent a useful tool to reproducibly study NASH progression. sPCLS can be used to evaluate potential treatments for NASH, as demonstrated in our elafibranor study, and serves as a model to bridge results from rodent studies to the human system.NEW & NOTEWORTHY This study showed that nonalcoholic steatohepatitis can be studied ex vivo in precision-cut liver slices obtained from murine diet-induced fatty livers. Liver slices develop a spontaneous inflammatory and fibrogenic response during culture that can be augmented with specific modulators. Additionally, the model can be used to test the efficacy of pharmaceutical compounds (as shown in this investigation with elafibranor) and could be a tool for preclinical assessment of potential therapies.


Subject(s)
Inflammation/pathology , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Cells, Cultured , Chalcones/pharmacology , Collagen Type I/biosynthesis , Diet , Disease Models, Animal , In Vitro Techniques , Lipid Metabolism/genetics , Lipopolysaccharides/pharmacology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Propionates/pharmacology , Transcriptome/genetics , Transforming Growth Factor beta1/pharmacology
18.
Toxicol Appl Pharmacol ; 391: 114916, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035996

ABSTRACT

Fibroblast-to-myofibroblast differentiation is one of the most important characteristics of pulmonary fibrosis, and screening natural compounds targeting fibroblast differentiation is always a promising approach to discover drug candidates for treatment of pulmonary fibrosis. Trehalose reportedly has many potential medical applications, especially in treating neurodegeneration diseases. However, it remains unclear whether trehalose suppresses lung fibroblast differentiation. In this work, we found that trehalose decreased the expression levels of α-smooth muscle actin (α-SMA) following the induction of transforming growth factor ß1 (TGF-ß1) in pretreatment, co-treatment, and post-treatment groups. Trehalose also reduced the production of type I collagen, lung fibroblast-containing gel contractility and cell filament formation in TGF-ß1-stimulated MRC-5 cells. Although trehalose is a known autophagy inducer, our results showed that its suppressive effect on fibroblast differentiation was not via trehalose-induced autophagy. And it did not affect canonical TGFß/Smad2/3 pathway. By applying proteomic profiling technology, we demonstrated that the downregulation of ß-catenin was involved in the trehalose-repressive action on fibroblast differentiation. The ß-catenin agonist, SKL2001, reversed the suppressive effect of trehalose on fibroblast differentiation. Overall, these experiments demonstrated that trehalose suppressed fibroblast differentiation via the downregulation of ß-catenin, but not through canonical autophagy and TGFß/Smad2/3 pathway, which is not only a novel understanding of trehalose, but also quite helpful for in vivo research of trehalose on pulmonary fibrosis in future.


Subject(s)
Cell Differentiation/drug effects , Fibroblasts/drug effects , Lung/cytology , Myofibroblasts/drug effects , Proteomics/methods , Transforming Growth Factor beta1/genetics , Trehalose/pharmacology , Actins/biosynthesis , Actins/genetics , Autophagy/drug effects , Cell Line , Collagen Type I/biosynthesis , Down-Regulation , Humans , Imidazoles/pharmacology , Isoxazoles/pharmacology , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Trehalose/antagonists & inhibitors , beta Catenin/agonists , beta Catenin/antagonists & inhibitors
19.
Lung ; 198(2): 275-282, 2020 04.
Article in English | MEDLINE | ID: mdl-32088750

ABSTRACT

PURPOSE: In obstructive sleep apnea (OSA), hypoxia secondary to apnea and hypopnea and the resulting systemic inflammatory response are the main causes of comorbidities. The aim of this study was to investigate the relationship between OSA and vimentin, which plays an important role in the activation of cells that synthesize inflammatory cytokines. MATERIALS AND METHODS: The study included 150 OSA patients (50 mild, 50 moderate, and 50 severe OSA) and 50 patients without OSA as a control group. Plasma vimentin levels were measured from peripheral blood samples using a commercial enzyme-linked immunosorbent assay (ELISA) kit. RESULTS: The OSA patients in our study had significantly higher body mass index, apnea-hypopnea index (AHI), triglyceride level, mean oxygen desaturation, and plasma vimentin levels compared to the healthy control group (p = 0.007, 0.001, 0004, 0.001, and 0.001, respectively). Plasma vimentin level was significantly higher in the moderate and severe OSA groups compared to the control and mild OSA groups (p = 0.001 for all). There was no difference between severe and moderate OSA. There were significant correlations between plasma vimentin levels and OSA patients' AHI and mean oxygen desaturation (r = 0.46, p = 0.001; r = 0.214, p = 0.005). CONCLUSION: In this study, we observed significant positive correlations between plasma vimentin level and OSA severity, weight, AHI, and mean oxygen desaturation. Vimentin may have utility as a biomarker in the follow-up and treatment of OSA.


Subject(s)
Collagen Type I/biosynthesis , Oxygen Consumption , Sleep Apnea, Obstructive , Vimentin/blood , Biomarkers/blood , Body Mass Index , Correlation of Data , Endothelial Cells/physiology , Female , Humans , Inflammation/metabolism , Macrophage Activation/physiology , Male , Middle Aged , Monitoring, Physiologic/methods , Polysomnography/methods , Severity of Illness Index , Sleep Apnea, Obstructive/blood , Sleep Apnea, Obstructive/etiology , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/therapy , Triglycerides/blood
20.
Pharmacology ; 105(7-8): 454-460, 2020.
Article in English | MEDLINE | ID: mdl-31812960

ABSTRACT

INTRODUCTION: Although it is beneficial to protect the skin from natural aging, especially in an aging society, the approach by which this can be achieved is still not well known. Hochu-ekki-to, a Chinese natural medicine, has various advantageous effects; however, there is no report about its influence on skin aging. OBJECTIVE: Therefore, we examined the effect of hochu-ekki-to against natural aging. METHODS: Hairless mice, bred without ultraviolet ray irradiation and physical stress, were orally administered huchu-ekki-to 3 times per week for 2 years. After that period, degree of skin hydration and permeability were measured. Furthermore, hematoxylin and eosin histochemistry was performed to determine the morphology and condition of the tissues. Lastly, levels of vitamin A, vitamin C, and reactive oxygen species (ROS) in plasma and skin, as well as concentration of hyaluronic acid in the skin, were measured. RESULTS: Signs of skin aging were ameliorated by administration of hochu-ekki-to, such as moisture retention, skin hydration, and the generation of wrinkles. Furthermore, vitamin A, vitamin C, collagen type I, collagen type III, fibroblasts, and hyaluronic acid levels in the skin increased, while levels of ROS decreased after hochu-ekki-to treatment. CONCLUSION: These results indicated that natural skin aging was ameliorated by treatment with hochu-ekki-to, specifically moisture retention, and skin hydration, and thickening, via the regulation of the vitamin C/fibroblast, collagen type III/collagen type I, and vitamin A/hyaluronic acid signaling pathways.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Skin Aging/drug effects , Skin/drug effects , Skin/metabolism , Animals , Ascorbic Acid/blood , Collagen Type I/biosynthesis , Collagen Type III/biosynthesis , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronic Acid/biosynthesis , Male , Mice , Mice, Hairless , Reactive Oxygen Species/metabolism , Vitamin A/blood
SELECTION OF CITATIONS
SEARCH DETAIL